Abstract
The endogenous opioid system is largely expressed in the brain, and both endogenous opioid peptides and receptors are present in areas associated with reward and motivation. It is well known that this endogenous system plays a key role in many aspects of addictive behaviours. The present review summarizes the modifications of the opioid system induced by chronic treatment with drugs of abuse reported in preclinical and clinical studies, as well as the action of opioid antagonists and agonists on the reinforcing effects of drugs of abuse, with therapeutic perspectives. We have focused on the effects of chronic psychostimulants, alcohol and nicotine exposure. Taken together, the changes in both opioid peptides and opioid receptors in different brain structures following acute or chronic exposure to these drugs of abuse clearly identify the opioid system as a potential target for the development of effective pharmacotherapy for the treatment of addiction and the prevention of relapse.
Tables of Links
TARGETS |
---|
δ receptor (DOP receptor) |
κ receptor (KOP receptor) |
μ receptor (MOP receptor) |
LIGANDS | |||
---|---|---|---|
5′-guanidinonaltrindol | CTOP | ICI-174,864 | Naloxone |
β-endorphin | Dexamphetamine | JDTic | Naltrexone |
Amphetamine | Diprenorphine | Leu-enkephalin | Naltrindole |
Bremazocine | Dopamine | Met-enkephalin | Nicotine |
Buprenorphine | Dynorphin | Methadone | Nor-BNI |
Cocaine | Ethanol | Methylphenidate | Prodynorphin |
CTAP | Glutamate | Nalmefene | U50,488 |
Glutamine | Naloxonazine |
These Tables list key protein targets and ligands in this article which are hyperlinked to corresponding entries in http://www.guidetopharmacology.org, the common portal for data from the IUPHAR/BPS Guide to PHARMACOLOGY (Pawson et al., 2014) and are permanently archived in the Concise Guide to PHARMACOLOGY 2013/14 (Alexander et al., 2013).
Introduction
Addiction is a worldwide public health problem for which there are currently no satisfactory treatments. While the existing medications permit efficient detoxification, methods for curing this condition are not yet available. Whatever the drugs of abuse used, a very high percentage of patients relapse into drug use, even after a long period of abstinence. There is an urgent need for new therapeutic strategies that should be based on our new understanding of the neurobiological mechanisms of addiction.
Drug addiction vulnerability is affected by a combination of genetic, epigenetic and environmental factors coupled with drug-induced effects. Neurochemical alterations in the brain caused by addictive drugs have a cellular and molecular basis and, in the setting of repeated self-exposure, leading to addiction, these changes may be persistent. However, the understanding of the cellular and biochemical mechanisms underlying both compulsive drug-seeking behaviour and the very long persistence of addictive effects is still very limited.
The brain network mediating the rewarding properties of drugs and craving phenomena has been identified. The system has been shown to involve such structures as the nucleus accumbens (NAc), ventral tegmental area (VTA), prefrontal cortex (PFC) and limbic structures, in particular the so-called extended amygdala. The enhancement of dopamine secretion in the NAc is a common effect of drugs of abuse. This effect can result from both a direct action on dopaminergic neurons (e.g. cocaine and amphetamine) and an indirect effect by modifying the activity of certain populations of neurons such as GABAergic interneurons that interact with dopaminergic VTA neurons (e.g. ethanol, opioids). The last 60 years of research has provided extraordinary advances in our knowledge of the reward system. For a long time, research on reward mechanisms traditionally focused on brain dopamine. However, from all the results reported in the literature using dopamine agonists or antagonists, it clearly appears that while dopamine plays a key role in reward, it is not the only neurotransmitter involved.
Research on the endogenous opioid system has substantially contributed to our understanding of the molecular mechanisms of drug addiction. Opioid receptors and their endogenous peptide ligands are largely distributed through the CNS and peripheral tissues. The existence of opioid receptors in the brain was demonstrated for the first time in 1973 by three independent groups (Pert and Snyder, 1973; Simon et al., 1973; Terenius, 1973), and it is only in the mid-1990s that the different opioid receptors were cloned: δ (DOP), μ (MOP) and κ (KOP) receptors (Evans et al., 1992; Kieffer et al., 1992; Chen et al., 1993; Li et al., 1993; Meng et al., 1993; Thompson et al., 1993; Yasuda et al., 1993). Three precursors of endogenous peptides have also been cloned, proopiomelanocortin (POMC), proenkephalin (PENK) or prodynorphin (PDYN). These precursors generate several final active peptides: β-endorphin from POMC, methionine-enkephalin (Met-enkephalin) and leucine-enkephalin (Leu-enkephalin) from PENK, dynorphins and neo-endorphins from PDYN. These endogenous opioid ligands exhibit different affinities for each opioid receptor. β-Endorphin shows a higher affinity for MOP receptors, Met- and Leu-enkephalin bind to DOP receptors with an affinity 20-fold greater than that for MOP receptors, and dynorphins are the endogenous ligands for KOP receptors (for review, see Corbett et al., 1993).
The different components of the endogenous opioid system are highly expressed in brain areas involved in reward and motivation (for review, see Mansour et al., 1995). Thus, opioid peptides and receptors are present in the VTA, NAc, PFC, hypothalamus and extended amygdala (Mansour et al., 1993; 1994a,b,; 1995; Delfs et al., 1994), and participate in the modulation of the reward circuits.
The main goal of this review is to describe the current knowledge concerning the contribution of the endogenous opioid system to the addictive properties of the different drugs of abuse. Considering the substantial amount of data from animal studies implicating the endogenous opioid system in reward and addiction, combined with the results from human post-mortem brains or obtained by neuroimaging in addicts, several questions about their clinical relevance arise: to what extent is the endogenous opioid system a valuable target for developing new treatments for the management of addiction? How successful are pharmacotherapies targeting the opioid system for the treatment of addiction? What could be the future directions?
This review gives an overview of how the opioid system is regulated by different drugs of abuse, to achieve a better knowledge regarding the use of opioid ligands in addiction treatments. With this aim, a specific focus has been done on psychostimulants (i.e. cocaine, amphetamine, methylphenidate), alcohol and nicotine. Opioid and cannabinoid addictions have not been considered as opioid ligands are already largely used in clinic to treat opioid addiction. Moreover, although numerous preclinical evidences indicate interplay between opioid and cannabinoid systems, only few studies have investigated the potential therapeutic interest of opioid ligands in cannabis addiction.
Cocaine
Modification of endogenous opioid system by psychostimulant treatment
Acute and chronic administration of psychostimulants produce adaptive changes in opioid peptide content, gene expression and receptor densities in brain structures related to reward circuits. However, one of the major difficulties is the discrepancies in the results reported in the literature, which is not surprising as it is now well established that neuroadaptations depend on different factors, including drug administration pattern and withdrawal period (see Conclusion section).
The most reliable finding in terms of psychostimulant-induced regulation of opioid peptide gene expression is the increase in PDYN mRNA and peptide levels in the striatum, but only shortly after chronic treatment (30 min, 1 h or 3 h after the last injection of cocaine or methylphenidate; Smiley et al., 1990; Steiner and Gerfen, 1993; Daunais and McGinty, 1995; Spangler et al., 1996a; Torres and Horowitz, 1999; Brandon and Steiner, 2003; Fagergren et al., 2003; Bailey et al., 2005), which is abolished by a selective antagonist or deletion of D1 receptors (Daunais and McGinty, 1996; Moratalla et al., 1996). A return to control levels was observed within 24 h (Smiley et al., 1990), and in animals treated for 10 days with cocaine followed by a 10-day drug free period, a decrease of PDYN mRNA was reported (Svensson and Hurd, 1998). Regarding dynorphin peptide levels, while a single dose of cocaine did not affect the level of dynorphin, a repeated treatment (4 days) increased striatal content of dynorphin, which persisted for at least 4 days and returned to the basal level 12 days after the last injection (Sivam, 1989). Using a microdialysis approach, methamphetamine was also shown to elevate extracellular levels of dynorphin peptide in the striatum, suggesting an increase in peptide release (Bustamante et al., 2002).
In other brain structures, preclinical studies provide conflicting data. For instance, repeated administration of cocaine or amphetamine (three injections per day for 5 days) was reported to induce a long-lasting increase in PDYN mRNA levels in the NAc (Turchan et al., 1998), whereas other studies (three injections per day for 10 or 14 days) failed to detect cocaine-induced changes in PDYN gene expression in the same nucleus (Daunais and McGinty, 1995; Mathieu-Kia and Besson, 1998; Romualdi et al., 2001).
Regarding changes in PENK mRNA following chronic cocaine treatment in rodents, the results are highly inconsistent whatever the brain structures studied and the specific pattern of drug administration, with increase (Branch et al., 1992; Hurd et al., 1992; Steiner and Gerfen, 1993; Przewlocka and Lason, 1995; Spangler et al., 1996b; Mathieu-Kia and Besson, 1998; Svensson and Hurd, 1998; Crespo et al., 2001; Zhang et al., 2012), or no change (Hurd et al., 1992; Daunais and McGinty, 1995; Mathieu-Kia and Besson, 1998; Alvarez Fischer et al., 2001; Bailey et al., 2005; Ziolkowska et al., 2006). Taken together, the results of the studies reviewed here suggest that the magnitude and significance of the changes in PENK gene expression are complex and clearly depend upon the brain region (e.g. NAc, hypothalamus, central amygdala, frontal cortex, olfactory tubercle), the type of drug administration (self-administration, repeated injections) and also the duration of the withdrawal period. A post-mortem study in humans with a history of cocaine abuse, reported a decrease in PENK mRNA in the caudate putamen, with a reduction in enkephalin peptide levels (Hurd and Herkenham, 1993).
The involvement of β-endorphin in the acquisition of cocaine self-administration has also been reported, with a transient increase in extracellular levels of β-endorphin in the NAc during cocaine self-administration (Roth-Deri et al., 2003; 2008,). The rewarding action of acute cocaine was reduced in β-endorphin-deficient mice (Nguyen et al., 2012). The release of β-endorphin in the NAc may function as a mechanism for lowering of cue-induced craving. However, this mechanism appears to be short-lived as 30 days after, cue exposure did not induce an increase in β-endorphin levels (Dikshtein et al., 2013).
The influence of cocaine on MOP, DOP and KOP receptor immunoreactivity and binding remains controversial, and changes are dynamic and vary according to the stage of the addiction cycle and brain regions (Azaryan et al., 1998; Bailey et al., 2007; Gorelick et al., 2008). In a post-mortem study, the density of KOP receptors in the NAc and other limbic brain regions was increased twofold in cocaine users as compared with control subjects (Staley et al., 1997). In a preclinical study, a decrease in KOP receptor density has been reported in the NAc only after chronic cocaine treatment, whereas this decrease was observed in the striatum after both acute and chronic injections (Turchan et al., 1998). This decrease in the striatum may reflect a compensatory down-regulation of KOP receptors in response to PDYN induction.
Neuroimaging of cocaine users using PET showed increased MOP receptor binding in several brain regions (e.g. frontal, lateral temporal, anterior cingulate cortex and amygdala) that correlated positively with cocaine craving and prevalence to relapse (Zubieta et al., 1996; Gorelick et al., 2005; Ghitza et al., 2010). These results are in agreement with those reported in preclinical studies. Thus, acute binge cocaine administration increased MOP receptor mRNA levels in the frontal cortex, NAc and amygdala, but not in the striatum, thalamus, hippocampus and hypothalamus (Yuferov et al., 1999). In addition a significant increase in the level of MOP receptor mRNA was detected in the NAc after 3 days of cocaine treatment with no modifications of DOP receptors in rats (Azaryan et al., 1996). In another study, DOP receptor mRNA levels were elevated in the VTA of rats expressing amphetamine behavioural sensitization after short-term withdrawal (2 days; Magendzo and Bustos, 2003).
Action of opioid antagonists or agonists on reinforcing effects of psychostimulants
MOP receptor antagonists were able to block the development of cocaine-induced behavioural sensitization, as well as the rewarding properties of cocaine, measured by the conditioned place preference (CPP) model. Given the evidence that MOP and dopamine receptors are co-localized within individual neurons of the striatum (Ambrose et al., 2004), it is not surprising that the blockade of MOP receptors could have profound effects on behaviours mediated in part by the striatal dopamine system. Thus, μ preferential opioid antagonists, naloxone and naltrexone, and μ-selective antagonists (CTOP, CTAP, naloxonazine) were able to reduce cocaine-induced CPP in rodents (Rademacher and Steinpreis, 2002; Schroeder et al., 2007). Moreover, MOP receptor antisense attenuated the expression of cocaine-induced behavioural sensitization and cocaine-induced CPP (Hummel et al., 2006). In MOP receptor knockout (KO) mice, cocaine-induced CPP was maintained (Contarino et al., 2002; Hall et al., 2004; Nguyen et al., 2012) or decreased (Hall et al., 2004) depending on the dose and experimental conditions (number and duration of conditioning sessions). These data suggest that activation of MOP receptors by endogenous opioid peptides subsequent to cocaine administration plays an important role in the subjective rewarding effects of cocaine and the development of cocaine-induced CPP.
Systemic administration of naloxone or naltrexone was able to reduce cocaine self-administration in rats (Corrigall and Coen, 1991; Giuliano et al., 2013), in good agreement with the results obtained in MOP receptor KO mice, where cocaine self-administration was reduced (Mathon et al., 2005). Other studies have also shown that, following their microinfusion in the VTA, a selective μ-opioid receptor antagonists (CTOP) produced a small decrease in cocaine self-administration (Corrigall et al., 1999), and a selective μ-agonist has been shown to enhance the reinforcing effects of the drug (Corrigall et al., 1999). No effect was found following naltrexone microinjection in the caudate, amygdala, NAc or medial PFC, while in the VTA, blockade of endogenous opioid receptors attenuated cocaine self-administration (Ramsey et al., 1999).
Strikingly, high doses of methadone were able to block the acquisition and expression of cocaine-induced CPP, and to interfere with incubation of cocaine sensitization and associated alterations in gene expression (Leri et al., 2012), while they did not alter self-administration (Leri et al., 2009). There is also evidence that buprenorphine can reduce cocaine use in patients with a history of i.v. cocaine, inhibit cocaine self-administration in rats, reduce cocaine seeking during extinction in the self-administration model, and block cocaine-induced sensitization (Kosten et al., 1991; Foltin and Fischman, 1996; Kuzmin et al., 2000; Sorge et al., 2005; Sorge and Stewart, 2006; Wee et al., 2012). It remains puzzling that buprenorphine reduces cocaine seeking; one hypothesis could be that buprenorphine is able to increase basal levels of glutamate in the NAc, which could contribute to its moderating effects on cocaine-induced effects (Placenza et al., 2008).
DOP receptors also seem to play an important role in the reinforcing effects of cocaine. Thus, naltrindole was able to significantly block cocaine-induced CPP, and inhibit cocaine self-administration (Menkens et al., 1992; Suzuki et al., 1994; Reid et al., 1995). These data further support the role of processes associated with DOP receptors in the ability of cocaine to reinforce its own use. However, other studies have shown that naltrindole, at doses that did not modify the locomotor activity of animals (0.03–3.0 mg·kg−1), did not alter the number of cocaine infusions taken by the rats in the self-administration paradigm, while a higher dose of naltrindole (10 mg·kg−1), which markedly depressed locomotor activity, resulted in a low (16%) reduction of cocaine self-administration behaviour (de Vries et al., 1995). In a more recent study, using brain microinjection, it has been demonstrated that naltrindole 5′-isothiocyanate decreased cocaine self-administration when injected into the NAc, but increased this behaviour when administered in the VTA (Ward and Roberts, 2007). Interestingly, administration of the δ-selective antagonist into the amygdala was without effect. This suggests that the modulation of cocaine rewarding effects by δ-opioid antagonists is brain region-dependent.
Several studies have investigated the role of κ-opioid ligands on the reinforcing effects of cocaine. Thus, it has been shown that κ-opioid agonists were able to reduce cocaine-induced CPP, cocaine self-administration and cocaine-induced decreases in intracerebral self-stimulation thresholds (Suzuki et al., 1992; Glick et al., 1995; Tomasiewicz et al., 2008), suggesting that activation of KOP receptors reduces the reward-related effects of cocaine. Regarding cocaine-induced behavioural sensitization, the results are controversial. While it has been shown that a single injection of the KOP receptor agonists attenuated the expression of cocaine-induced behavioural sensitization in rats (Collins et al., 2001; Morani et al., 2012), it has also been reported that the κ-opioid antagonist nor-BNI blocked cocaine locomotor sensitization, but in a model of food restriction in rats (Allen et al., 2013). The results obtained with the κ-antagonists remain highly controversial. While some authors suggest that these antagonists are able to reduce cocaine self-administration (Kuzmin et al., 1998), others show that they produced either no effects or small effects that did not show consistent trends with doses (Corrigall et al., 1999). Furthermore, blockade of KOP receptors attenuated the development of depressive-like behaviours induced by cocaine withdrawal in rats (Chartoff et al., 2012).
Therapeutic perspectives
Effective medications to treat cocaine dependence have not been identified. Numerous studies have pointed out a role for endogenous opioid systems in behavioural effects induced by cocaine (see also for recent reviews, Yoo et al., 2012; Charbogne et al., 2014). Overall, these findings suggest that endogenous opioid transmission facilitates cocaine-influenced behaviour and that MOP and KOP receptors may represent specific target sites for therapeutic or behavioural intervention related to cocaine addiction. Mixed κ- and μ-ligands have been developed, with either agonist or antagonist properties, which are able to decrease cocaine self-administration in rats (Archer et al., 1996; Glick et al., 1998; Neumeyer et al., 2001). The use of buprenorphine in preclinical studies consistently induced a reduction in cocaine self-administration (Mello et al., 1989; Carroll and Lac, 1992). In the clinic, the efficacy of buprenorphine in reducing cocaine use among opiate-dependent subjects has been demonstrated (Mendelson et al., 1992; Strain et al., 1994; Foltin and Fischman, 1996; Kouri et al., 1996; Montoya et al., 2004), but with differences in subject characteristics (e.g. differences in cocaine use or in comorbid psychiatric disorders) or differences in study methods that may affect treatment outcome. Buprenorphine together with naltrexone is being investigated as a potential combination treatment in response to the need to expand treatment options for cocaine dependence (Mooney et al., 2013). Interestingly, similar results to those obtained with buprenorphine were obtained with methadone (Strain et al., 1994; Foltin and Fischman, 1996).
The effects of naltrexone on the subjective and physiological effects of amphetamine were also investigated, using dexamphetamine as a model substance in patients diagnosed with amphetamine dependence (Jayaram-Lindstrom et al., 2008). This study was performed on a small homogeneous population of male amphetamine-dependent patients, and needs to be extended. However, the results clearly demonstrated that naltrexone attenuated the subjective effects and the craving for dexamphetamine.
Alcohol
Modification of endogenous opioid system by alcohol treatment
Many data have demonstrated a change in the opioid system (peptides and receptors) upon acute or chronic ethanol treatment. Similar to cocaine, the most consistent effect of alcohol on opioid peptides is an increase in dynorphin in reward-related brain structures. Indeed, the dynorphin level (mRNA or protein) was increased in the NAc (Przewlocka et al., 1994; Lindholm et al., 2000) and amygdala (D'Addario et al., 2013b), a stress-related brain area, following chronic ethanol exposure. This increase, especially after a protracted withdrawal would contribute to the negative effects of ethanol withdrawal (Gillett et al., 2013). Recent data suggested that dynorphin up-regulation by alcohol could be caused by epigenetic modifications (D'Addario et al., 2013a). Data are scarce concerning the influence of ethanol on opioid peptides in humans but they seemed to match with those obtained in animals. Thus, in post-mortem human brains of alcoholics, PDYN mRNA was increased in the dorsolateral PFC (Bazov et al., 2013).
The results with enkephalins are more controversial, as an increase, decrease or no change have been observed whatever the duration of ethanol exposure (acute or chronic), type of administration (contingent or non-contingent), the withdrawal period or the brain structure considered (Schulz et al., 1980; Seizinger et al., 1983; Przewlocka et al., 1997; Lindholm et al., 2000; Marinelli et al., 2005; Mendez and Morales-Mulia, 2006; Jarjour et al., 2009). For instance, following an acute injection of ethanol, the Met-enkephalin level was increased in the NAc (Seizinger et al., 1983; Marinelli et al., 2005; Mendez et al., 2010) but not in the VTA (Jarjour et al., 2009). In humans, no change in PENK mRNA was detected in any brain structures tested (dorsolateral PFC, orbitofrontal cortex and hippocampus; Bazov et al., 2013). This result supports a minor role of enkephalins in ethanol addiction as evidenced by the ability of enkephalin KO mice to still prefer ethanol in a two-bottle choice (Koenig and Olive, 2002) and self-administration paradigms (Hayward et al., 2004).
There are few data on the effects of ethanol exposure on β-endorphin in reward-related brain structures. However, most of the data demonstrates that acute ethanol promotes an increase in β-endorphin, and chronic ethanol had mixed effects (increase, decrease or no changes). Using microdialysis, Gianoulakis and co-workers found an increase in β-endorphin after an acute injection of ethanol in the central amygdala and VTA (Lam et al., 2008; Jarjour et al., 2009) that could involve corticotropin-releasing hormone receptors (Lam and Gianoulakis, 2011). In rats continuously exposed in a one-bottle access situation to ethanol, no variation in β-endorphin was measured in the NAc or VTA (Leriche and Mendez, 2010). However, in a protocol of voluntary alcohol ingestion using the two-bottle choice paradigm, POMC mRNA was increased in the NAc Shell (Zhou et al., 2013). In humans the β-endorphin neuropeptide level was augmented in blood during ethanol intoxication (Barret et al., 1987; Aguirre et al., 1995b), whereas it was diminished during withdrawal (Aguirre et al., 1990; 1995a,) contributing to anxiety (Kiefer et al., 2002).
Chronic ethanol exposure might lead to different effects on opioid receptors with opposite results on DOP and MOP receptors. In rats with free access to an ethanol-containing liquid diet, immunohistochemistry analysis revealed a decrease in MOP receptors in NAc, cortex and hippocampus whereas DOP receptors were decreased in the hippocampus (Saland et al., 2005). Interestingly, after acute exposure using an intra-gastric injection, the number of MOP receptors in the striatum was not modified (Mendez et al., 2003; Leriche and Mendez, 2010). With regard to the KOP receptors, a recent study found a transient increase (only observed 30 min after the last injection, but no later) of its coding mRNA in the amygdala after a 5 day treatment of intra-gastric administration of ethanol (D'Addario et al., 2013b).
Using [11C]-diprenorphine (a non-selective opioid receptor ligand), Williams and co-workers found an increase in opioid receptor availability in the early abstinence period of ethanol-dependent patients and a positive correlation between [11C]-diprenorphine volume distribution and craving (Williams et al., 2009). The same correlation was found with MOP receptors using a selective ligand, [11C]-carfentanil. Indeed, an increase in MOP receptors was found in detoxified patients and correlated with the severity of alcohol craving (Heinz et al., 2005). Apparent opposite results were found by Bencherif and co-workers, where a lower MOP receptor binding potential in some sub-regions of the cortex was associated with a higher craving in alcohol-dependent subjects (Bencherif et al., 2004). This discrepancy might be explained by the different structures analyzed as Heinz and collaborators observed these changes in the ventral striatum, a structure not tested in Bencherif's study.
Action of opioid antagonists or agonists on reinforcing effects of alcohol
Numerous papers have been published on the role of opioid receptors in ethanol intake and reinforcing effects. Using naloxone, it was shown that the blockade of opioid receptors reduced ethanol intake and preference in Sprague-Dawley rats (Reid and Hunter, 1984; Barson et al., 2009) or in rats selectively bred for high ethanol preference (Froehlich et al., 1990).
The three opioid receptors were individually tested for their role in ethanol addiction. In MOP receptor KO mice, no operant self-administration of ethanol was observed and the two-bottle choice test even revealed an aversion for alcohol (Roberts et al., 2000). Microinjection of CTAP revealed that the NAc and ventral pallidum are important regions for these MOP receptor-mediated effects on ethanol consumption (Perry and McNally, 2013a,b,). The VTA is also important as a knockdown of MOP receptors in this region with small hairpin-RNA reduced ethanol intake in the two-bottle choice paradigm in mice (Lasek et al., 2007). However, results for the involvement of the DOP receptor are less consistent. DOP receptor KO mice showed a preference for ethanol measured in the two-bottle choice paradigm and have an increased ethanol intake in this same test only after operant ethanol administration (Roberts et al., 2001). In contrast, other authors found that ICI-174,864 and naltrindole, two DOP-selective antagonists, reduce ethanol intake in the two-bottle choice test in rats selectively bred for ethanol preference (Krishnan-Sarin et al., 1995), whereas other authors did not observe such results in regular (not ethanol preferring) rats (Stromberg et al., 1998) or rhesus monkeys (Williams and Woods, 1998). KOP receptor agonists such as U50,488H (Lindholm et al., 2001) or bremazocine (Nestby et al., 1999) decreased ethanol intake in the two-bottle choice paradigm. U50,488H has also been found to block acquisition of ethanol in CPP (Logrip et al., 2009). According to these data, nor-BNI had no effects on ethanol intake (Holter et al., 2000) or operant self-administration (Doyon et al., 2006) in naive animals. However, in certain conditions, the role of KOP receptors in ethanol addiction switches. Indeed, in a rat strain, selectively bred for alcohol preference, or in ethanol-dependent animals, KOP antagonists reduce operant self-administration (Walker and Koob, 2008; Kissler et al., 2014; Rorick-Kehn et al., 2014). Taken together, these findings indicate that whereas KOP receptor activation reduces ethanol reinforcement in non-dependent animals probably via an aversive effect, the κ-opioid system may participate in ethanol seeking in dependent subjects (Wee and Koob, 2010).
Therapeutic perspectives
Preclinical data strongly suggest that blocking opioid receptors might be helpful in reducing some characteristics of ethanol addiction. Naltrexone, a preferential MOP receptor antagonist that is able to bind to other opioid receptors at higher concentrations (Raynor et al., 1994; Wang et al., 2007), was the first drug acting on opioid receptors approved for the treatment of ethanol dependence and the second drug specific for the treatment of this condition 40 years after disulfiram. Naltrexone has been shown to reduce ethanol intake in the two-bottle choice paradigm (Stromberg et al., 1998; Parkes and Sinclair, 2000) and inhibit operant self-administration of alcohol in rodents (Bienkowski et al., 1999; Walker and Koob, 2008). Naltrexone is rapidly absorbed when taken orally and is converted into several metabolites including 6β-naltrexol, the main metabolite, which has been shown to reduce ethanol drinking in rodents (Stromberg et al., 2002). Using [11C]-carfentanil, a PET study demonstrated that 50 mg of naltrexone (corresponding to the daily dose) block 90% of brain MOP receptors after 48 h (Lee et al., 1988) explaining its long-term action. Studies have also shown that oral administration of naltrexone for a few weeks reduces craving (Chick et al., 2000) and prevents relapse (Morris et al., 2001). However, its efficacy fades over time as demonstrated by studies investigating long-term treatment (Balldin et al., 2003; Krystal et al, 2001). Interestingly, some factors that contribute to a naltrexone-positive response have been identified, such as the MOP receptor single-nucleotide polymorphism A118G (Oslin et al., 2003; Anton et al., 2008) and adherence to treatment (Chick et al., 2000; Krystal et al, 2001). This could explain the differences in the ratio between responding versus non-responding patients among clinical studies. To avoid a lack of adherence to treatment, an injectable extended-release formula of naltrexone has been developed and has been shown to be effective at blocking MOP receptors in rats for 1 month (Bartus et al., 2003). It seems to be well-tolerated and promoted reductions in heavy drinking among treatment-seeking alcohol-dependent patients during 6 months of therapy (Garbutt et al., 2005). More interestingly, it improved their quality of life, specifically in numerous domains such as mental health and social functioning (Pettinati et al., 2009).
Nalmefene, a naltrexone analogue, has been approved by regulatory agencies in the treatment of alcohol dependence. Nalmefene differs from naltrexone by the presence of a methylene group instead of the ketone at the 6-position, which increases affinity towards opioid receptors (Emmerson et al., 1994), and half-life (10 h; Dixon et al., 1987). In a preclinical study, nalmefene reduced operant self-administration of ethanol in rats (June et al., 1998). With the exception of one study (Anton et al., 2004), clinical trials have demonstrated the efficacy of nalmefene in treating ethanol dependence (Mason et al., 1999; Karhuvaara et al., 2007; Gual et al., 2013; Mann et al., 2013), with a reduced relapse rate to heavy drinking when this treatment was combined with cognitive behavioural therapy (Mason et al., 1999). Recently, a new opioid receptor antagonist has been released, LY2196044 (WO 2004/026305) and presents promising results in increasing the abstinence period in ethanol-dependent, treatment-seeking patients (Wong et al., 2014).
Nicotine
Modification of endogenous opioid system by nicotine treatment
As with cocaine and alcohol exposures, tobacco smoking induces functional alterations in the endogenous opioid system. Because nicotine is considered as the main active component responsible for the addictive properties of tobacco, numerous studies have focused on the effects of chronic nicotine administration on the endogenous opioid system in various regions of the brain (for review, see Berrendero et al., 2010; Drews and Zimmer, 2010; Hadjiconstantinou and Neff, 2011). Chronic exposure to nicotine alters the release of endogenous opioid peptides in the brain and those alterations are specific to the nature of the endogenous opioid peptide being investigated. In addition, these modifications are persistent, dynamic and time-specific [e.g. it depends when the measure is done during nicotine treatment, or when the measure is performed after nicotine withdrawal (early vs. late)].
With regard to the effects on dynorphin synthesis and release induced by chronic nicotine administration, a decrease in dynorphin content was observed in the mice striatum from 30 min to 72 h after the last nicotine injection (Isola et al., 2008). A compensatory mechanism involving opioid synthesis is also implemented, in which PDYN mRNA was increased in the same reward-related brain structure from 8 h to 96 h after the last injection. However, no change was observed in the biosynthesis and release of dynorphin in rat striatum (Hollt and Horn, 1992; Mathieu et al., 1996; Mathieu-Kia and Besson, 1998), indicating the importance of nicotine dose, treatment schedule and species for the observed changes.
The effects of chronic nicotine treatment on Met-enkephalin and PENK have been extensively investigated by many research groups. PENK mRNA was decreased in the striatum of mice 2 h following nicotine cessation followed by a rebound increase lasting for over 72 h (Houdi et al., 1998). Similarly, following 14 days of chronic nicotine treatment, Met-enkephalin levels were decreased in the rat striatum after 1 h of nicotine cessation (Wewers et al., 1999). PENK mRNA was increased in striatum and NAc 24 h after the last injection of a chronic nicotine treatment in rats (Mathieu et al., 1996) and mice (Dhatt et al., 1995) but not 2 h after nicotine cessation in rats (Mathieu-Kia and Besson, 1998). From 4 to over 72 h after nicotine cessation, Met-enkephalin levels and PENK mRNA were increased in the NAc (Isola et al., 2002). Overall, these findings highlight a biphasic change in the levels of Met-enkephalin and PENK mRNA in striatum, with a decrease during early withdrawal and an increase during a more protracted withdrawal period. This biphasic response may reflect alterations in the synthesis and metabolism of Met-enkephalin.
Chronic nicotine treatment was shown to have a biphasic effect on the hypothalamic β-endorphin level in mice (Rosecrans et al., 1985). Chronic nicotine exposure induced first a decrease in hypothalamic β-endorphin levels 24 h after the last injection (Gudehithlu et al., 2012). Within 7 days, the β-endorphin levels returned to the baseline and even increased above the baseline after 14 days of nicotine withdrawal. In contrast, another study reported a long-lasting inhibition of POMC gene expression in the mediobasohypothalamus (Rasmussen, 1998). It seems that chronic nicotine diminishes the synthesis of β-endorphin in the limbic areas (e.g. striatum, hippocampus, hypothalamus, PFC) that might contribute to aversive states associated with nicotine withdrawal (Berrendero et al., 2010; Drews and Zimmer, 2010; Hadjiconstantinou and Neff, 2011; Gudehithlu et al., 2012). However, the situation is far less clear with the nicotine-induced release of β-endorphin if we consider the clinical studies. In current smokers, their levels of peripheral plasma β-endorphin have been found to be increased (Backon, 1989; del Arbol et al., 2000; Gilbert et al., 1992; Pomerleau et al., 1983; Seyler et al., 1986). This apparent discrepancy between animal and human findings may result from the lack of a direct relationship between peripheral and central β-endorphin levels (Berrendero et al., 2010).
Several studies have investigated the effects of chronic nicotine treatment on the densities, affinities and functional activities of MOP, DOP and KOP receptors. It has been shown that a 14 day treatment with nicotine induces an up-regulation of MOP receptors in rat striatum (Wewers et al., 1999). Chronic nicotine administration decreased the density of MOP receptors in the striatum and NAc in C57BL/6 mice (Galeote et al., 2006) but not in NMRI mice (Vihavainen et al., 2008). However, both the affinity and the functional activity of MOP receptors were unchanged by the chronic treatment in these two strains (Galeote et al., 2006; Vihavainen et al., 2008). Finally, an uncoupling and desensitization of KOP and DOP receptors in the striatum and NAc were observed during nicotine withdrawal, whereas the densities of these receptors were unaltered (McCarthy et al., 2010; 2011,).
In humans, using [11C]-carfentanil a down-regulation of MOP receptors in the thalamus, ventral basal ganglia and amygdala has been reported after smoking nicotine cigarettes (Scott et al., 2007; Weerts et al., 2014). In addition, a basal reduction in MOP receptor availability in different brain structures (caudate, cingulate, globus pallidus, insula, putamen, thalamus and ventral striatum) has been reported to be negatively correlated to severity of nicotine dependence (Weerts et al., 2014).
Action of opioid antagonists and agonists on reinforcing effects of nicotine
Pharmacological and genetic approaches in preclinical studies have provided evidence for the involvement of the endogenous opioid system in nicotine-rewarding effects (for review, see Berrendero et al., 2010; Maldonado, 2010; Charbogne et al., 2014). The MOP receptor is particularly involved in nicotine-rewarding effects and nicotine withdrawal. Thus, administration of the glycyl-glutamine, a MOP receptor antagonist, inhibited the acquisition and the expression of nicotine-induced CPP and attenuated withdrawal signs in rats (Goktalay et al., 2006). The preferential MOP antagonist naloxone abolished nicotine-induced CPP (Zarrindast et al., 2003; Walters et al., 2005), attenuated nicotine-induced conditioned place aversion in mice (Zarrindast et al., 2003) and decreased nicotine self-administration in rats (Ismayilova and Shoaib, 2010; but see also, Corrigall and Coen, 1991). Activation of MOP receptors is required for the reinforcement of nicotine in rats, as shown by the reduction of nicotine self-administration in rats pretreated with the selective MOP receptor antagonist naloxonazine (Liu and Jernigan, 2011). In addition, naltrexone, another preferential MOP antagonist, was able to attenuate nicotine cue-maintained responding during extinction and cue-induced reinstatement of nicotine-seeking behaviour after extinction in a self-administration test, suggesting that MOP antagonists would be good candidates for the prevention of smoking relapse triggered by exposure to environmental smoking cues (Liu et al., 2009).
Studies using genetically modified mice have confirmed the crucial role of MOP receptors in the rewarding effects of nicotine. Indeed, nicotine-induced CPP was attenuated in mice lacking MOP receptors, PENK or β-endorphin (Berrendero et al., 2002; 2005,; Trigo et al., 2009). These findings strongly suggest that activation of MOP receptors by endogenous enkephalins and β-endorphins are required to obtain the reinforcing effects of nicotine (Berrendero et al., 2010).
A KO study suggested that DOP/PENK signalling also contributed to the reinforcing effects of nicotine (Berrendero et al., 2012). In this study, DOP receptor KO mice did not express a nicotine-induced CPP and displayed a lower percentage of acquisition of i.v. nicotine self-administration. This result has been confirmed by a decrease in the rate of acquisition in wild type mice pretreated with the DOP receptor antagonist naltrindole. However, other pharmacological studies failed to reveal an effect of naltrindole on nicotine self-administration in rats (Ismayilova and Shoaib, 2010; Liu and Jernigan, 2011).
KOP receptor agonist and antagonist studies are difficult to interpret and how KOP receptor activity influences nicotine reinforcement needs to be investigated further. For example, the KOP agonist U50,488 has revealed a dual role of the endogenous κ-opioid system on nicotine self-administration with a decrease in nicotine intake at the high dose of agonist and a trend for an increase with a lower dose in rats (Ismayilova and Shoaib, 2010), suggesting that κ-agonists bind with lower affinity to other receptors, activation of which produces opposing effects to those resulting from the activation of a higher affinity binding site. Nevertheless, using 5′-guanidinonaltrindole, a selective KOP antagonist, Liu and Jernigan reported a lack of involvement of KOP receptor activation by dynorphin on nicotine self-administration (Liu and Jernigan, 2011). In addition, the selective KOP antagonist JDTic failed to block the expression of nicotine reward in the CPP paradigm (Jackson et al., 2010), supporting a role for the KOP/dynorphin system in mediating dysphoric aspects during withdrawal rather than the reinforcing properties of nicotine. This contribution of dynorphin to aversive effects of nicotine has been supported by a recent study in PDYN KO mice, showing a decrease in self-administration of a low dose of nicotine (Galeote et al., 2009).
Therapeutic perspectives
The high prevalence of smoking among heroin addicts (Mello et al., 1980) and methadone- or buprenorphine-maintained patients (Chait and Griffiths, 1984; Mello et al., 1985; Mutschler et al., 2002; Zirakzadeh et al., 2013) highlights interactions between the opioid and nicotine systems that may lead to an increase in the reinforcing effects of smoking. Because of the critical role of MOP receptors in the reinforcing effects of nicotine, one of the therapeutic strategies available is to attenuate the rewarding effects of cigarette smoking by opioid antagonists. Thus, numerous clinical trials have been performed to evaluate the effect of preferential μ-opioid antagonists, naloxone and naltrexone, on smoking cessation (see David et al., 2013 for review). However, it seems that there were no or weak overall effects of naloxone alone or in association with nicotine replacement therapy on long-term smoking abstinence (Karras and Kane, 1980; Nemeth-Coslett and Griffiths, 1986; Gorelick et al., 1988). Because naloxone displays a short duration of action, a lot of recent studies focused on naltrexone, an opioid antagonist with longer acting effects, but the same inconclusive results have been obtained (Covey et al., 1999; Wong et al., 1999; Ahmadi et al., 2003; Krishnan-Sarin et al., 2003; O'Malley et al., 2006; Toll et al., 2010; David et al., 2013). Moreover, evaluation of naltrexone effects in preventing nicotine relapse and craving did not result in a clear picture (Wewers et al., 1998; Hutchison et al., 1999; Krishnan-Sarin et al., 2003; Rohsenow et al., 2007). Compared with placebo, significant trends towards a cessation at the end of naltrexone treatments were recorded among patients; however, these positive effects did not seem long lasting, differences between both groups were attenuated at 6 months (Covey et al., 1999). Interestingly, naltrexone in combination with nicotine replacement therapy resulted in an increase in abstinence rates (Krishnan-Sarin et al., 2003) and prevented weight gain following smoking cessation (Krishnan-Sarin et al., 2003; O'Malley et al., 2006).
Conclusion
In the past decades, many advances in our understanding of the underlying biology of addiction have opened the doors to the development of novel pharmacotherapies. As reported in this review, both endogenous opioid peptides and receptors play a key role in many aspects of addictive behaviours. It clearly appears that drugs of abuse modify the activity of the endogenous opioid system, and produce adaptive changes that play important roles in the development/maintenance of addiction. Moreover, the modifications of endogenous opioid systems induced by drugs of abuse are dynamic processes and vary according to the stage of the addiction cycle. These dynamic changes should be taken into consideration, and may explain why clinical trials, using pharmacotherapies to treat addiction, report modest efficacy, or describe efficient results only on a sub-population of patients (Potenza et al., 2011; Volkow and Skolnick, 2012).
This review shows that opioid ligands may be very useful, whatever the drug of abuse used, as the endogenous opioid system is a common neurobiological substrate for certain components of addictive processes induced by the different drugs of abuse. Nevertheless, it clearly appears throughout the review that the data in the literature are not consistent. Regarding the regulation of endogenous opioid system by psychostimulants, alcohol or nicotine, numerous protocols have been used (treatment duration, withdrawal period, pattern of administration, strains of animals …) that may explain the divergence, as it is now well established that neuroadaptations depend on different factors. The time after drug of abuse administration appears to be a determining factor for detecting increased gene expression. Moreover, numerous brain structures are heterogeneous (e.g. cortex, striatum, amygdala, NAc), and discrete changes within the larger structures may be missed, or different results may be observed in subregions. Some technical approaches do not allow us to discriminate between these subregions (e.g. Western blot), and/or the precise areas of analyses in most papers are not described, pointing out the limitations of the methods that may explain the differences in the results reported. Similarly, several discrepancies are reported in the literature regarding the action of opioid ligands on the reinforcing effects of psychostimulants, alcohol and nicotine. Several animal models are used to investigate different components of drug addiction, with different protocols. Animal models of substance abuse include both non-contingent (experimenter-administered) and contingent (self-administered) drug administration. A simple animal model to study the rewarding effect of drugs, CPP uses a classical Pavlovian conditioning procedure to pair an unconditioned stimulus (e.g. cocaine) with a designated area and measure the preference for the stimulus-paired area compared with the unpaired area. An increase in preference for the stimulus-paired area serves as a measure of its Pavlovian rewarding effects (e.g. Bardo and Bevins, 2000; Tzschentke, 2007). Another model, behavioural sensitization, is defined as a progressive enhancement of drug-induced responses that develops during repeated drug treatment and then persists even after weeks of withdrawal. It can be produced by exposure to either contingent or non-contingent drugs of abuse. The induction of sensitization involves brain structures common to those known to play a role in reward processes, and it is considered to be a good marker of neurochemical changes that underlie addiction (e.g. (Vanderschuren and Kalivas, 2000). The third model largely used in preclinical studies is the self-administration paradigm that refers to training rodents in an operant chamber to press a lever or poke their nose in a hole in order to receive an i.v. infusion of drug (e.g. Ahmed, 2012). Self-administration procedures can differ in many ways, including whether training to respond for food precedes drug self-administration, the dose of drug available, the number of responses required to obtain the drug, and the daily duration of drug access. Clinical studies using opioid ligands also report some discrepancies. However, they are generally conducted on a small or on a heterogeneous population of patients. In addition, the end points defined in the clinical trials to measure the effects of a treatment and how this treatment may improve health status are often different, which makes it difficult to compare results across studies. Whatever the reasons behind the variable results, overall, the potential of opioid ligands as a pharmaceutical treatment for psychostimulants, alcohol and nicotine is promising and merits further investigation.
Another therapeutic approach that could be helpful in the future is the enkephalin-degrading enzyme inhibitors, as it has been suggested that such inhibitors could represent effective treatments for addiction (Roques and Noble, 1996; Noble and Roques, 2003). These inhibitors could be used alone or in association with positive allosteric modulators (Burford et al., 2013; 2014; 2015,), which have the specific advantage of only modulating the activity of the receptor when the orthosteric site binds an endogenous agonist, thus maintaining spatial and temporal control of receptor signalling in vivo. These allosteric modulators have little or no detectable functional activity when bound to the receptor in the absence of an orthosteric agonist, but can potentiate the activity of bound orthosteric agonist, seen as an increase in apparent potency and/or efficacy of the orthosteric agonist.
However, in order for an effective treatment to prevent the relapses in addicted patients, pharmacotherapies must be associated with structured psychosocial therapies to enhance strategies to prevent relapse and encourage compliance with treatment. Moreover, a consideration of any genetic variants of opioid receptors is important when determining treatment options for different individuals, and possibly crucial in determining which patients are likely to respond to opioid ligand treatment (Oslin et al., 2006; Sturgess et al., 2011; Thorsell, 2013; Garbutt et al., 2014). Benefits of such an approach, in addition to increasing treatment response and health, may increase the cost-effectiveness of a treatment, as well as decreasing the risk of exposing individuals to medication that is ineffective.
Author contributions
F. N., L. M. and N. M. managed the literature searches, and contributed to write the first draft of the manuscript. F. N. organized the first draft and prepared the final version of the manuscript. All the authors contributed to and have approved the final manuscript.
Conflict of interest
The authors declare they have no conflict of interest.
Glossary
- CPP
conditioned place preference
- DOP receptor
δ-opioid receptor
- KO
knockout
- KOP receptor
κ-opioid receptor
- Leu-enkephalin
leucine-enkephalin
- Met-enkephalin
methionine-enkephalin
- MOP receptor
μ-opioid receptor
- NAc
nucleus accumbens
- PDYN
prodynorphin
- PENK
proenkephalin
- PFC
prefrontal cortex
- POMC
proopiomelanocortin
- VTA
ventral tegmental area
References
- Aguirre JC, Del Arbol JL, Raya J, Ruiz-Requena ME, Rico Irles J. Plasma beta-endorphin levels in chronic alcoholics. Alcohol. 1990;7:409–412. doi: 10.1016/0741-8329(90)90024-7. [DOI] [PubMed] [Google Scholar]
- Aguirre JC, del Arbol JL, Rico J, Raya J, Miranda MT. Classification of alcoholics on the basis of plasma beta-endorphin concentration. Alcohol. 1995a;12:531–534. doi: 10.1016/0741-8329(95)00039-9. [DOI] [PubMed] [Google Scholar]
- Aguirre JC, del Arbol JL, Rico J, Raya J, Ruiz-Requena ME. Effect of acute alcohol intoxication on the opioid system in humans. Alcohol. 1995b;12:559–562. doi: 10.1016/0741-8329(95)02002-0. [DOI] [PubMed] [Google Scholar]
- Ahmadi J, Ashkani H, Ahmadi M, Ahmadi N. Twenty-four week maintenance treatment of cigarette smoking with nicotine gum, clonidine and naltrexone. J Subst Abuse Treat. 2003;24:251–255. doi: 10.1016/s0740-5472(03)00027-8. [DOI] [PubMed] [Google Scholar]
- Ahmed SH. The science of making drug-addicted animals. Neuroscience. 2012;211:107–125. doi: 10.1016/j.neuroscience.2011.08.014. [DOI] [PubMed] [Google Scholar]
- Alexander SPH, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Spedding M, et al. The Concise Guide to PHARMACOLOGY 2013/14: G protein-coupled receptors. Br J Pharmacol. 2013;170:1459–1581. doi: 10.1111/bph.12445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allen CP, Zhou Y, Leri F. Effect of food restriction on cocaine locomotor sensitization in Sprague-Dawley rats: role of kappa opioid receptors. Psychopharmacology (Berl) 2013;226:571–578. doi: 10.1007/s00213-012-2930-7. [DOI] [PubMed] [Google Scholar]
- Alvarez Fischer D, Schafer MK, Ferger B, Gross S, Westermann R, Weihe E, et al. Sensitization to the behavioural effects of cocaine: alterations in tyrosine hydroxylase or endogenous opioid mRNAs are not necessarily involved. Naunyn Schmiedebergs Arch Pharmacol. 2001;363:288–294. doi: 10.1007/s002100000332. [DOI] [PubMed] [Google Scholar]
- Ambrose LM, Unterwald EM, Van Bockstaele EJ. Ultrastructural evidence for co-localization of dopamine D2 and micro-opioid receptors in the rat dorsolateral striatum. Anat Rec A Discov Mol Cell Evol Biol. 2004;279:583–591. doi: 10.1002/ar.a.20054. [DOI] [PubMed] [Google Scholar]
- Anton RF, Pettinati H, Zweben A, Kranzler HR, Johnson B, Bohn MJ, et al. A multi-site dose ranging study of nalmefene in the treatment of alcohol dependence. J Clin Psychopharmacol. 2004;24:421–428. doi: 10.1097/01.jcp.0000130555.63254.73. [DOI] [PubMed] [Google Scholar]
- Anton RF, Oroszi G, O'Malley S, Couper D, Swift R, Pettinati H, et al. An evaluation of mu-opioid receptor (OPRM1) as a predictor of naltrexone response in the treatment of alcohol dependence: results from the Combined Pharmacotherapies and Behavioral Interventions for Alcohol Dependence (COMBINE) study. Arch Gen Psychiatry. 2008;65:135–144. doi: 10.1001/archpsyc.65.2.135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- del Arbol JL, Munoz JR, Ojeda L, Cascales AL, Irles JR, Miranda MT, et al. Plasma concentrations of beta-endorphin in smokers who consume different numbers of cigarettes per day. Pharmacol Biochem Behav. 2000;67:25–28. doi: 10.1016/s0091-3057(00)00291-4. [DOI] [PubMed] [Google Scholar]
- Archer S, Glick SD, Bidlack JM. Cyclazocine revisited. Neurochem Res. 1996;21:1369–1373. doi: 10.1007/BF02532378. [DOI] [PubMed] [Google Scholar]
- Azaryan AV, Coughlin LJ, Buzas B, Clock BJ, Cox BM. Effect of chronic cocaine treatment on mu- and delta-opioid receptor mRNA levels in dopaminergically innervated brain regions. J Neurochem. 1996;66:443–448. doi: 10.1046/j.1471-4159.1996.66020443.x. [DOI] [PubMed] [Google Scholar]
- Azaryan AV, Clock BJ, Rosenberger JG, Cox BM. Transient upregulation of mu opioid receptor mRNA levels in nucleus accumbens during chronic cocaine administration. Can J Physiol Pharmacol. 1998;76:278–283. [PubMed] [Google Scholar]
- Backon J. Negative correlation of cigarette smoking and dysmenorrhea: reduced prostaglandin synthesis due to beta-endorphin, nicotine, or acrolein antagonism. Med Hypotheses. 1989;28:213–214. doi: 10.1016/0306-9877(89)90054-6. [DOI] [PubMed] [Google Scholar]
- Bailey A, Yuferov V, Bendor J, Schlussman SD, Zhou Y, Ho A, et al. Immediate withdrawal from chronic ‘binge’ cocaine administration increases mu-opioid receptor mRNA levels in rat frontal cortex. Brain Res Mol Brain Res. 2005;137:258–262. doi: 10.1016/j.molbrainres.2005.02.017. [DOI] [PubMed] [Google Scholar]
- Bailey A, Gianotti R, Ho A, Kreek MJ. Downregulation of kappa-opioid receptors in basolateral amygdala and septum of rats withdrawn for 14 days from an escalating dose ‘binge’ cocaine administration paradigm. Synapse. 2007;61:820–826. doi: 10.1002/syn.20436. [DOI] [PubMed] [Google Scholar]
- Balldin J, Berglund M, Borg S, Mansson M, Bendtsen P, Franck J, et al. A 6-month controlled naltrexone study: combined effect with cognitive behavioral therapy in outpatient treatment of alcohol dependence. Alcohol Clin Exp Res. 2003;27:1142–1149. doi: 10.1097/01.ALC.0000075548.83053.A9. [DOI] [PubMed] [Google Scholar]
- Bardo MT, Bevins RA. Conditioned place preference: what does it add to our preclinical understanding of drug reward? Psychopharmacology (Berl) 2000;153:31–43. doi: 10.1007/s002130000569. [DOI] [PubMed] [Google Scholar]
- Barret L, Bourhis F, Buffet H, Danel V, Debru JL. Determination of beta-endorphin in alcoholic patients in the acute stage of intoxication: relation with naloxone therapy. Drug Alcohol Depend. 1987;19:71–78. doi: 10.1016/0376-8716(87)90088-3. [DOI] [PubMed] [Google Scholar]
- Barson JR, Carr AJ, Soun JE, Sobhani NC, Leibowitz SF, Hoebel BG. Opioids in the nucleus accumbens stimulate ethanol intake. Physiol Behav. 2009;98:453–459. doi: 10.1016/j.physbeh.2009.07.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bartus RT, Emerich DF, Hotz J, Blaustein M, Dean RL, Perdomo B, et al. Vivitrex, an injectable, extended-release formulation of naltrexone, provides pharmacokinetic and pharmacodynamic evidence of efficacy for 1 month in rats. Neuropsychopharmacology. 2003;28:1973–1982. doi: 10.1038/sj.npp.1300274. [DOI] [PubMed] [Google Scholar]
- Bazov I, Kononenko O, Watanabe H, Kuntic V, Sarkisyan D, Taqi MM, et al. The endogenous opioid system in human alcoholics: molecular adaptations in brain areas involved in cognitive control of addiction. Addict Biol. 2013;18:161–169. doi: 10.1111/j.1369-1600.2011.00366.x. [DOI] [PubMed] [Google Scholar]
- Bencherif B, Wand GS, McCaul ME, Kim YK, Ilgin N, Dannals RF, et al. Mu-opioid receptor binding measured by [11C]carfentanil positron emission tomography is related to craving and mood in alcohol dependence. Biol Psychiatry. 2004;55:255–262. doi: 10.1016/j.biopsych.2003.07.007. [DOI] [PubMed] [Google Scholar]
- Berrendero F, Kieffer BL, Maldonado R. Attenuation of nicotine-induced antinociception, rewarding effects, and dependence in mu-opioid receptor knock-out mice. J Neurosci. 2002;22:10935–10940. doi: 10.1523/JNEUROSCI.22-24-10935.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berrendero F, Mendizabal V, Robledo P, Galeote L, Bilkei-Gorzo A, Zimmer A, et al. Nicotine-induced antinociception, rewarding effects, and physical dependence are decreased in mice lacking the preproenkephalin gene. J Neurosci. 2005;25:1103–1112. doi: 10.1523/JNEUROSCI.3008-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berrendero F, Robledo P, Trigo JM, Martin-Garcia E, Maldonado R. Neurobiological mechanisms involved in nicotine dependence and reward: participation of the endogenous opioid system. Neurosci Biobehav Rev. 2010;35:220–231. doi: 10.1016/j.neubiorev.2010.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berrendero F, Plaza-Zabala A, Galeote L, Flores A, Bura SA, Kieffer BL, et al. Influence of delta-opioid receptors in the behavioral effects of nicotine. Neuropsychopharmacology. 2012;37:2332–2344. doi: 10.1038/npp.2012.88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bienkowski P, Kostowski W, Koros E. Ethanol-reinforced behaviour in the rat: effects of naltrexone. Eur J Pharmacol. 1999;374:321–327. doi: 10.1016/s0014-2999(99)00245-9. [DOI] [PubMed] [Google Scholar]
- Branch AD, Unterwald EM, Lee SE, Kreek MJ. Quantitation of preproenkephalin mRNA levels in brain regions from male Fischer rats following chronic cocaine treatment using a recently developed solution hybridization assay. Brain Res Mol Brain Res. 1992;14:231–238. doi: 10.1016/0169-328x(92)90178-e. [DOI] [PubMed] [Google Scholar]
- Brandon CL, Steiner H. Repeated methylphenidate treatment in adolescent rats alters gene regulation in the striatum. Eur J Neurosci. 2003;18:1584–1592. doi: 10.1046/j.1460-9568.2003.02892.x. [DOI] [PubMed] [Google Scholar]
- Burford NT, Clark MJ, Wehrman TS, Gerritz SW, Banks M, O'Connell J, et al. Discovery of positive allosteric modulators and silent allosteric modulators of the mu-opioid receptor. Proc Natl Acad Sci U S A. 2013;110:10830–10835. doi: 10.1073/pnas.1300393110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burford NT, Wehrman T, Bassoni D, O'Connell J, Banks M, Zhang L, et al. Identification of selective agonists and positive allosteric modulators for micro- and delta-opioid receptors from a single high-throughput screen. J Biomol Screen. 2014;19:1255–1265. doi: 10.1177/1087057114542975. [DOI] [PubMed] [Google Scholar]
- Burford NT, Traynor JR, Alt A. Positive allosteric modulators of the mu-opioid receptor: a novel approach for future pain medications. Br J Pharmacol. 2015;172:277–286. doi: 10.1111/bph.12599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bustamante D, You ZB, Castel MN, Johansson S, Goiny M, Terenius L, et al. Effect of single and repeated methamphetamine treatment on neurotransmitter release in substantia nigra and neostriatum of the rat. J Neurochem. 2002;83:645–654. doi: 10.1046/j.1471-4159.2002.01171.x. [DOI] [PubMed] [Google Scholar]
- Carroll ME, Lac ST. Effects of buprenorphine on self-administration of cocaine and a nondrug reinforcer in rats. Psychopharmacology (Berl) 1992;106:439–446. doi: 10.1007/BF02244812. [DOI] [PubMed] [Google Scholar]
- Chait LD, Griffiths RR. Effects of methadone on human cigarette smoking and subjective ratings. J Pharmacol Exp Ther. 1984;229:636–640. [PubMed] [Google Scholar]
- Charbogne P, Kieffer BL, Befort K. 15 years of genetic approaches in vivo for addiction research: opioid receptor and peptide gene knockout in mouse models of drug abuse. Neuropharmacology. 2014;76(Pt B):204–217. doi: 10.1016/j.neuropharm.2013.08.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chartoff E, Sawyer A, Rachlin A, Potter D, Pliakas A, Carlezon WA. Blockade of kappa opioid receptors attenuates the development of depressive-like behaviors induced by cocaine withdrawal in rats. Neuropharmacology. 2012;62:167–176. doi: 10.1016/j.neuropharm.2011.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Mestek A, Liu J, Hurley JA, Yu L. Molecular cloning and functional expression of a mu-opioid receptor from rat brain. Mol Pharmacol. 1993;44:8–12. [PubMed] [Google Scholar]
- Chick J, Anton R, Checinski K, Croop R, Drummond DC, Farmer R, et al. A multicentre, randomized, double-blind, placebo-controlled trial of naltrexone in the treatment of alcohol dependence or abuse. Alcohol Alcohol. 2000;35:587–593. doi: 10.1093/alcalc/35.6.587. [DOI] [PubMed] [Google Scholar]
- Collins SL, D'Addario C, Izenwasser S. Effects of kappa-opioid receptor agonists on long-term cocaine use and dopamine neurotransmission. Eur J Pharmacol. 2001;426:25–34. doi: 10.1016/s0014-2999(01)01194-3. [DOI] [PubMed] [Google Scholar]
- Contarino A, Picetti R, Matthes HW, Koob GF, Kieffer BL, Gold LH. Lack of reward and locomotor stimulation induced by heroin in mu-opioid receptor-deficient mice. Eur J Pharmacol. 2002;446:103–109. doi: 10.1016/s0014-2999(02)01812-5. [DOI] [PubMed] [Google Scholar]
- Corbett AD, Paterson SJ, Kosterlitz HW. Selectivity of ligands for opioid receptors. In: Herz A, editor. Opioids. Vol. 104. Berlin Heidelberg: Springer-Verlag; 1993. pp. 645–679. Vol. [Google Scholar]
- Corrigall WA, Coen KM. Opiate antagonists reduce cocaine but not nicotine self-administration. Psychopharmacology (Berl) 1991;104:167–170. doi: 10.1007/BF02244173. [DOI] [PubMed] [Google Scholar]
- Corrigall WA, Coen KM, Adamson KL, Chow BL. The mu opioid agonist DAMGO alters the intravenous self-administration of cocaine in rats: mechanisms in the ventral tegmental area. Psychopharmacology (Berl) 1999;141:428–435. doi: 10.1007/s002130050853. [DOI] [PubMed] [Google Scholar]
- Covey LS, Glassman AH, Stetner F. Naltrexone effects on short-term and long-term smoking cessation. J Addict Dis. 1999;18:31–40. doi: 10.1300/J069v18n01_04. [DOI] [PubMed] [Google Scholar]
- Crespo JA, Manzanares J, Oliva JM, Corchero J, Palomo T, Ambrosio E. Extinction of cocaine self-administration produces a differential time-related regulation of proenkephalin gene expression in rat brain. Neuropsychopharmacology. 2001;25:185–194. doi: 10.1016/S0893-133X(01)00221-4. [DOI] [PubMed] [Google Scholar]
- Daunais JB, McGinty JF. Cocaine binges differentially alter striatal preprodynorphin and zif/268 mRNAs. Brain Res Mol Brain Res. 1995;29:201–210. doi: 10.1016/0169-328x(94)00246-b. [DOI] [PubMed] [Google Scholar]
- Daunais JB, McGinty JF. The effects of D1 or D2 dopamine receptor blockade on zif/268 and preprodynorphin gene expression in rat forebrain following a short-term cocaine binge. Brain Res Mol Brain Res. 1996;35:237–248. doi: 10.1016/0169-328x(95)00226-i. [DOI] [PubMed] [Google Scholar]
- David SP, Lancaster T, Stead LF, Evins AE, Prochaska JJ. Opioid antagonists for smoking cessation. Cochrane Database Syst Rev. 2013;(6) doi: 10.1002/14651858.CD003086.pub3. CD003086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- D'Addario C, Caputi FF, Ekstrom TJ, Di Benedetto M, Maccarrone M, Romualdi P, et al. Ethanol induces epigenetic modulation of prodynorphin and pronociceptin gene expression in the rat amygdala complex. J Mol Neurosci. 2013a;49:312–319. doi: 10.1007/s12031-012-9829-y. [DOI] [PubMed] [Google Scholar]
- D'Addario C, Caputi FF, Rimondini R, Gandolfi O, Del Borrello E, Candeletti S, et al. Different alcohol exposures induce selective alterations on the expression of dynorphin and nociceptin systems related genes in rat brain. Addict Biol. 2013b;18:425–433. doi: 10.1111/j.1369-1600.2011.00326.x. [DOI] [PubMed] [Google Scholar]
- Delfs JM, Kong H, Mestek A, Chen Y, Yu L, Reisine T, et al. Expression of mu opioid receptor mRNA in rat brain: an in situ hybridization study at the single cell level. J Comp Neurol. 1994;345:46–68. doi: 10.1002/cne.903450104. [DOI] [PubMed] [Google Scholar]
- Dhatt RK, Gudehithlu KP, Wemlinger TA, Tejwani GA, Neff NH, Hadjiconstantinou M. Preproenkephalin mRNA and methionine-enkephalin content are increased in mouse striatum after treatment with nicotine. J Neurochem. 1995;64:1878–1883. doi: 10.1046/j.1471-4159.1995.64041878.x. [DOI] [PubMed] [Google Scholar]
- Dikshtein Y, Barnea R, Kronfeld N, Lax E, Roth-Deri I, Friedman A, et al. beta-endorphin via the delta opioid receptor is a major factor in the incubation of cocaine craving. Neuropsychopharmacology. 2013;38:2508–2514. doi: 10.1038/npp.2013.155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dixon R, Gentile J, Hsu HB, Hsiao J, Howes J, Garg D, et al. Nalmefene: safety and kinetics after single and multiple oral doses of a new opioid antagonist. J Clin Pharmacol. 1987;27:233–239. doi: 10.1002/j.1552-4604.1987.tb02191.x. [DOI] [PubMed] [Google Scholar]
- Doyon WM, Howard EC, Shippenberg TS, Gonzales RA. Kappa-opioid receptor modulation of accumbal dopamine concentration during operant ethanol self-administration. Neuropharmacology. 2006;51:487–496. doi: 10.1016/j.neuropharm.2006.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drews E, Zimmer A. Modulation of alcohol and nicotine responses through the endogenous opioid system. Prog Neurobiol. 2010;90:1–15. doi: 10.1016/j.pneurobio.2009.09.004. [DOI] [PubMed] [Google Scholar]
- Emmerson PJ, Liu MR, Woods JH, Medzihradsky F. Binding affinity and selectivity of opioids at mu, delta and kappa receptors in monkey brain membranes. J Pharmacol Exp Ther. 1994;271:1630–1637. [PubMed] [Google Scholar]
- Evans CJ, Keith DE, Jr, Morrison H, Magendzo K, Edwards RH. Cloning of a delta opioid receptor by functional expression. Science. 1992;258:1952–1955. doi: 10.1126/science.1335167. [DOI] [PubMed] [Google Scholar]
- Fagergren P, Smith HR, Daunais JB, Nader MA, Porrino LJ, Hurd YL. Temporal upregulation of prodynorphin mRNA in the primate striatum after cocaine self-administration. Eur J Neurosci. 2003;17:2212–2218. doi: 10.1046/j.1460-9568.2003.02636.x. [DOI] [PubMed] [Google Scholar]
- Foltin RW, Fischman MW. Effects of methadone or buprenorphine maintenance on the subjective and reinforcing effects of intravenous cocaine in humans. J Pharmacol Exp Ther. 1996;278:1153–1164. [PubMed] [Google Scholar]
- Froehlich JC, Harts J, Lumeng L, Li TK. Naloxone attenuates voluntary ethanol intake in rats selectively bred for high ethanol preference. Pharmacol Biochem Behav. 1990;35:385–390. doi: 10.1016/0091-3057(90)90174-g. [DOI] [PubMed] [Google Scholar]
- Galeote L, Kieffer BL, Maldonado R, Berrendero F. Mu-opioid receptors are involved in the tolerance to nicotine antinociception. J Neurochem. 2006;97:416–423. doi: 10.1111/j.1471-4159.2006.03751.x. [DOI] [PubMed] [Google Scholar]
- Galeote L, Berrendero F, Bura SA, Zimmer A, Maldonado R. Prodynorphin gene disruption increases the sensitivity to nicotine self-administration in mice. Int J Neuropsychopharmacol. 2009;12:615–625. doi: 10.1017/S1461145708009450. [DOI] [PubMed] [Google Scholar]
- Garbutt JC, Kranzler HR, O'Malley SS, Gastfriend DR, Pettinati HM, Silverman BL, et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a randomized controlled trial. JAMA. 2005;293:1617–1625. doi: 10.1001/jama.293.13.1617. [DOI] [PubMed] [Google Scholar]
- Garbutt JC, Greenblatt AM, West SL, Morgan LC, Kampov-Polevoy A, Jordan HS, et al. Clinical and biological moderators of response to naltrexone in alcohol dependence: a systematic review of the evidence. Addiction. 2014;109:1274–1284. doi: 10.1111/add.12557. [DOI] [PubMed] [Google Scholar]
- Ghitza UE, Preston KL, Epstein DH, Kuwabara H, Endres CJ, Bencherif B, et al. Brain mu-opioid receptor binding predicts treatment outcome in cocaine-abusing outpatients. Biol Psychiatry. 2010;68:697–703. doi: 10.1016/j.biopsych.2010.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilbert DG, Meliska CJ, Williams CL, Jensen RA. Subjective correlates of cigarette-smoking-induced elevations of peripheral beta-endorphin and cortisol. Psychopharmacology (Berl) 1992;106:275–281. doi: 10.1007/BF02801984. [DOI] [PubMed] [Google Scholar]
- Gillett K, Harshberger E, Valdez GR. Protracted withdrawal from ethanol and enhanced responsiveness stress: regulation via the dynorphin/kappa opioid receptor system. Alcohol. 2013;47:359–365. doi: 10.1016/j.alcohol.2013.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Giuliano C, Robbins TW, Wille DR, Bullmore ET, Everitt BJ. Attenuation of cocaine and heroin seeking by mu-opioid receptor antagonism. Psychopharmacology (Berl) 2013;227:137–147. doi: 10.1007/s00213-012-2949-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Glick SD, Maisonneuve IM, Raucci J, Archer S. Kappa opioid inhibition of morphine and cocaine self-administration in rats. Brain Res. 1995;681:147–152. doi: 10.1016/0006-8993(95)00306-b. [DOI] [PubMed] [Google Scholar]
- Glick SD, Visker KE, Maisonneuve IM. Effects of cyclazocine on cocaine self-administration in rats. Eur J Pharmacol. 1998;357:9–14. doi: 10.1016/s0014-2999(98)00548-2. [DOI] [PubMed] [Google Scholar]
- Goktalay G, Cavun S, Levendusky MC, Hamilton JR, Millington WR. Glycyl-glutamine inhibits nicotine conditioned place preference and withdrawal. Eur J Pharmacol. 2006;530:95–102. doi: 10.1016/j.ejphar.2005.11.034. [DOI] [PubMed] [Google Scholar]
- Gorelick DA, Rose J, Jarvik ME. Effect of naloxone on cigarette smoking. J Subst Abuse. 1988;1:153–159. doi: 10.1016/s0899-3289(88)80018-x. [DOI] [PubMed] [Google Scholar]
- Gorelick DA, Kim YK, Bencherif B, Boyd SJ, Nelson R, Copersino M, et al. Imaging brain mu-opioid receptors in abstinent cocaine users: time course and relation to cocaine craving. Biol Psychiatry. 2005;57:1573–1582. doi: 10.1016/j.biopsych.2005.02.026. [DOI] [PubMed] [Google Scholar]
- Gorelick DA, Kim YK, Bencherif B, Boyd SJ, Nelson R, Copersino ML, et al. Brain mu-opioid receptor binding: relationship to relapse to cocaine use after monitored abstinence. Psychopharmacology (Berl) 2008;200:475–486. doi: 10.1007/s00213-008-1225-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gual A, He Y, Torup L, van den Brink W, Mann K, Group ES. A randomised, double-blind, placebo-controlled, efficacy study of nalmefene, as-needed use, in patients with alcohol dependence. Eur Neuropsychopharmacol. 2013;23:1432–1442. doi: 10.1016/j.euroneuro.2013.02.006. [DOI] [PubMed] [Google Scholar]
- Gudehithlu KP, Duchemin AM, Tejwani GA, Neff NH, Hadjiconstantinou M. Nicotine-induced changes of brain beta-endorphin. Neuropeptides. 2012;46:125–131. doi: 10.1016/j.npep.2012.03.001. [DOI] [PubMed] [Google Scholar]
- Hadjiconstantinou M, Neff NH. Nicotine and endogenous opioids: neurochemical and pharmacological evidence. Neuropharmacology. 2011;60:1209–1220. doi: 10.1016/j.neuropharm.2010.11.010. [DOI] [PubMed] [Google Scholar]
- Hall FS, Goeb M, Li XF, Sora I, Uhl GR. mu-Opioid receptor knockout mice display reduced cocaine conditioned place preference but enhanced sensitization of cocaine-induced locomotion. Brain Res Mol Brain Res. 2004;121:123–130. doi: 10.1016/j.molbrainres.2003.10.024. [DOI] [PubMed] [Google Scholar]
- Hayward MD, Hansen ST, Pintar JE, Low MJ. Operant self-administration of ethanol in C57BL/6 mice lacking beta-endorphin and enkephalin. Pharmacol Biochem Behav. 2004;79:171–181. doi: 10.1016/j.pbb.2004.07.002. [DOI] [PubMed] [Google Scholar]
- Heinz A, Reimold M, Wrase J, Hermann D, Croissant B, Mundle G, et al. Correlation of stable elevations in striatal mu-opioid receptor availability in detoxified alcoholic patients with alcohol craving: a positron emission tomography study using carbon 11-labeled carfentanil. Arch Gen Psychiatry. 2005;62:57–64. doi: 10.1001/archpsyc.62.1.57. [DOI] [PubMed] [Google Scholar]
- Hollt V, Horn G. Effect of nicotine on mRNA levels encoding opioid peptides, vasopressin and alpha 3 nicotinic receptor subunit in the rat. Clin Investig. 1992;70:224–231. doi: 10.1007/BF00184655. [DOI] [PubMed] [Google Scholar]
- Holter SM, Henniger MS, Lipkowski AW, Spanagel R. Kappa-opioid receptors and relapse-like drinking in long-term ethanol-experienced rats. Psychopharmacology (Berl) 2000;153:93–102. doi: 10.1007/s002130000601. [DOI] [PubMed] [Google Scholar]
- Houdi AA, Dasgupta R, Kindy MS. Effect of nicotine use and withdrawal on brain preproenkephalin A mRNA. Brain Res. 1998;799:257–263. doi: 10.1016/s0006-8993(98)00454-5. [DOI] [PubMed] [Google Scholar]
- Hummel M, Schroeder J, Liu-Chen LY, Cowan A, Unterwald EM. An antisense oligodeoxynucleotide to the mu opioid receptor attenuates cocaine-induced behavioral sensitization and reward in mice. Neuroscience. 2006;142:481–491. doi: 10.1016/j.neuroscience.2006.06.013. [DOI] [PubMed] [Google Scholar]
- Hurd YL, Herkenham M. Molecular alterations in the neostriatum of human cocaine addicts. Synapse. 1993;13:357–369. doi: 10.1002/syn.890130408. [DOI] [PubMed] [Google Scholar]
- Hurd YL, Brown EE, Finlay JM, Fibiger HC, Gerfen CR. Cocaine self-administration differentially alters mRNA expression of striatal peptides. Brain Res Mol Brain Res. 1992;13:165–170. doi: 10.1016/0169-328x(92)90058-j. [DOI] [PubMed] [Google Scholar]
- Hutchison KE, Monti PM, Rohsenow DJ, Swift RM, Colby SM, Gnys M, et al. Effects of naltrexone with nicotine replacement on smoking cue reactivity: preliminary results. Psychopharmacology (Berl) 1999;142:139–143. doi: 10.1007/s002130050872. [DOI] [PubMed] [Google Scholar]
- Ismayilova N, Shoaib M. Alteration of intravenous nicotine self-administration by opioid receptor agonist and antagonists in rats. Psychopharmacology (Berl) 2010;210:211–220. doi: 10.1007/s00213-010-1845-4. [DOI] [PubMed] [Google Scholar]
- Isola R, Zhang H, Duchemin AM, Tejwani GA, Neff NH, Hadjiconstantinou M. Met-enkephalin and preproenkephalin mRNA changes in the striatum of the nicotine abstinence mouse. Neurosci Lett. 2002;325:67–71. doi: 10.1016/s0304-3940(02)00240-9. [DOI] [PubMed] [Google Scholar]
- Isola R, Zhang H, Tejwani GA, Neff NH, Hadjiconstantinou M. Dynorphin and prodynorphin mRNA changes in the striatum during nicotine withdrawal. Synapse. 2008;62:448–455. doi: 10.1002/syn.20515. [DOI] [PubMed] [Google Scholar]
- Jackson KJ, Carroll FI, Negus SS, Damaj MI. Effect of the selective kappa-opioid receptor antagonist JDTic on nicotine antinociception, reward, and withdrawal in the mouse. Psychopharmacology (Berl) 2010;210:285–294. doi: 10.1007/s00213-010-1803-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jarjour S, Bai L, Gianoulakis C. Effect of acute ethanol administration on the release of opioid peptides from the midbrain including the ventral tegmental area. Alcohol Clin Exp Res. 2009;33:1033–1043. doi: 10.1111/j.1530-0277.2009.00924.x. [DOI] [PubMed] [Google Scholar]
- Jayaram-Lindstrom N, Konstenius M, Eksborg S, Beck O, Hammarberg A, Franck J. Naltrexone attenuates the subjective effects of amphetamine in patients with amphetamine dependence. Neuropsychopharmacology. 2008;33:1856–1863. doi: 10.1038/sj.npp.1301572. [DOI] [PubMed] [Google Scholar]
- June HL, Grey C, Warren-Reese C, Durr LF, Ricks-Cord A, Johnson A, et al. The opioid receptor antagonist nalmefene reduces responding maintained by ethanol presentation: preclinical studies in ethanol-preferring and outbred Wistar rats. Alcohol Clin Exp Res. 1998;22:2174–2185. [PubMed] [Google Scholar]
- Karhuvaara S, Simojoki K, Virta A, Rosberg M, Loyttyniemi E, Nurminen T, et al. Targeted nalmefene with simple medical management in the treatment of heavy drinkers: a randomized double-blind placebo-controlled multicenter study. Alcohol Clin Exp Res. 2007;31:1179–1187. doi: 10.1111/j.1530-0277.2007.00401.x. [DOI] [PubMed] [Google Scholar]
- Karras A, Kane JM. Naloxone reduces cigarette smoking. Life Sci. 1980;27:1541–1545. doi: 10.1016/0024-3205(80)90562-7. [DOI] [PubMed] [Google Scholar]
- Kiefer F, Horntrich M, Jahn H, Wiedemann K. Is withdrawal-induced anxiety in alcoholism based on beta-endorphin deficiency? Psychopharmacology (Berl) 2002;162:433–437. doi: 10.1007/s00213-002-1118-y. [DOI] [PubMed] [Google Scholar]
- Kieffer BL, Befort K, Gaveriaux-Ruff C, Hirth CG. The delta-opioid receptor: isolation of a cDNA by expression cloning and pharmacological characterization. Proc Natl Acad Sci U S A. 1992;89:12048–12052. doi: 10.1073/pnas.89.24.12048. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kissler JL, Sirohi S, Reis DJ, Jansen HT, Quock RM, Smith DG, et al. The one-two punch of alcoholism: role of central amygdala dynorphins/kappa-opioid receptors. Biol Psychiatry. 2014;75:774–782. doi: 10.1016/j.biopsych.2013.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koenig HN, Olive MF. Ethanol consumption patterns and conditioned place preference in mice lacking preproenkephalin. Neurosci Lett. 2002;325:75–78. doi: 10.1016/s0304-3940(02)00242-2. [DOI] [PubMed] [Google Scholar]
- Kosten TA, Marby DW, Nestler EJ. Cocaine conditioned place preference is attenuated by chronic buprenorphine treatment. Life Sci. 1991;49:PL201–PL206. doi: 10.1016/0024-3205(91)90490-3. [DOI] [PubMed] [Google Scholar]
- Kouri EM, Lukas SE, Mendelson JH. P300 assessment of opiate and cocaine users: effects of detoxification and buprenorphine treatment. Biol Psychiatry. 1996;40:617–628. doi: 10.1016/0006-3223(95)00468-8. [DOI] [PubMed] [Google Scholar]
- Krishnan-Sarin S, Jing SL, Kurtz DL, Zweifel M, Portoghese PS, Li TK, et al. The delta opioid receptor antagonist naltrindole attenuates both alcohol and saccharin intake in rats selectively bred for alcohol preference. Psychopharmacology (Berl) 1995;120:177–185. doi: 10.1007/BF02246191. [DOI] [PubMed] [Google Scholar]
- Krishnan-Sarin S, Meandzija B, O'Malley S. Naltrexone and nicotine patch smoking cessation: a preliminary study. Nicotine Tob Res. 2003;5:851–857. doi: 10.1080/14622200310001614601. [DOI] [PubMed] [Google Scholar]
- Krystal JH, Cramer JA, Krol WF, Kirk GF, Rosenheck RA Veterans Affairs Naltrexone Cooperative Study G. Naltrexone in the treatment of alcohol dependence. N Engl J Med. 2001;345:1734–1739. doi: 10.1056/NEJMoa011127. [DOI] [PubMed] [Google Scholar]
- Kuzmin AV, Gerrits MA, Van Ree JM. Kappa-opioid receptor blockade with nor-binaltorphimine modulates cocaine self-administration in drug-naive rats. Eur J Pharmacol. 1998;358:197–202. doi: 10.1016/s0014-2999(98)00637-2. [DOI] [PubMed] [Google Scholar]
- Kuzmin AV, Gerrits MA, Zvartau EE, van Ree JM. Influence of buprenorphine, butorphanol and nalbuphine on the initiation of intravenous cocaine self-administration in drug naive mice. Eur Neuropsychopharmacol. 2000;10:447–454. doi: 10.1016/s0924-977x(00)00117-6. [DOI] [PubMed] [Google Scholar]
- Lam MP, Gianoulakis C. Effects of acute ethanol on corticotropin-releasing hormone and beta-endorphin systems at the level of the rat central amygdala. Psychopharmacology (Berl) 2011;218:229–239. doi: 10.1007/s00213-011-2337-x. [DOI] [PubMed] [Google Scholar]
- Lam MP, Marinelli PW, Bai L, Gianoulakis C. Effects of acute ethanol on opioid peptide release in the central amygdala: an in vivo microdialysis study. Psychopharmacology (Berl) 2008;201:261–271. doi: 10.1007/s00213-008-1267-8. [DOI] [PubMed] [Google Scholar]
- Lasek AW, Janak PH, He L, Whistler JL, Heberlein U. Downregulation of mu opioid receptor by RNA interference in the ventral tegmental area reduces ethanol consumption in mice. Genes Brain Behav. 2007;6:728–735. doi: 10.1111/j.1601-183X.2007.00303.x. [DOI] [PubMed] [Google Scholar]
- Lee MC, Wagner HN, Jr, Tanada S, Frost JJ, Bice AN, Dannals RF. Duration of occupancy of opiate receptors by naltrexone. J Nucl Med. 1988;29:1207–1211. [PubMed] [Google Scholar]
- Leri F, Zhou Y, Goddard B, Levy A, Jacklin D, Kreek MJ. Steady-state methadone blocks cocaine seeking and cocaine-induced gene expression alterations in the rat brain. Eur Neuropsychopharmacol. 2009;19:238–249. doi: 10.1016/j.euroneuro.2008.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leri F, Zhou Y, Carmichael B, Cummins E, Kreek MJ. Treatment-like steady-state methadone in rats interferes with incubation of cocaine sensitization and associated alterations in gene expression. Eur Neuropsychopharmacol. 2012;22:143–152. doi: 10.1016/j.euroneuro.2011.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leriche M, Mendez M. Ethanol exposure selectively alters beta-endorphin content but not [3H]-DAMGO binding in discrete regions of the rat brain. Neuropeptides. 2010;44:9–16. doi: 10.1016/j.npep.2009.11.009. [DOI] [PubMed] [Google Scholar]
- Li S, Zhu J, Chen C, Chen YW, Deriel JK, Ashby B, et al. Molecular cloning and expression of a rat kappa opioid receptor. Biochem J. 1993;295(Pt 3):629–633. doi: 10.1042/bj2950629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindholm S, Ploj K, Franck J, Nylander I. Repeated ethanol administration induces short- and long-term changes in enkephalin and dynorphin tissue concentrations in rat brain. Alcohol. 2000;22:165–171. doi: 10.1016/s0741-8329(00)00118-x. [DOI] [PubMed] [Google Scholar]
- Lindholm S, Werme M, Brene S, Franck J. The selective kappa-opioid receptor agonist U50,488H attenuates voluntary ethanol intake in the rat. Behav Brain Res. 2001;120:137–146. doi: 10.1016/s0166-4328(00)00368-5. [DOI] [PubMed] [Google Scholar]
- Liu X, Jernigan C. Activation of the opioid mu1, but not delta or kappa, receptors is required for nicotine reinforcement in a rat model of drug self-administration. Prog Neuropsychopharmacol Biol Psychiatry. 2011;35:146–153. doi: 10.1016/j.pnpbp.2010.10.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liu X, Palmatier MI, Caggiula AR, Sved AF, Donny EC, Gharib M, et al. Naltrexone attenuation of conditioned but not primary reinforcement of nicotine in rats. Psychopharmacology (Berl) 2009;202:589–598. doi: 10.1007/s00213-008-1335-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Logrip ML, Janak PH, Ron D. Blockade of ethanol reward by the kappa opioid receptor agonist U50,488H. Alcohol. 2009;43:359–365. doi: 10.1016/j.alcohol.2009.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magendzo K, Bustos G. Expression of amphetamine-induced behavioral sensitization after short- and long-term withdrawal periods: participation of mu- and delta-opioid receptors. Neuropsychopharmacology. 2003;28:468–477. doi: 10.1038/sj.npp.1300063. [DOI] [PubMed] [Google Scholar]
- Maldonado R. The endogenous opioid system and drug addiction. Ann Pharm Fr. 2010;68:3–11. doi: 10.1016/j.pharma.2009.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mann K, Bladstrom A, Torup L, Gual A, van den Brink W. Extending the treatment options in alcohol dependence: a randomized controlled study of as-needed nalmefene. Biol Psychiatry. 2013;73:706–713. doi: 10.1016/j.biopsych.2012.10.020. [DOI] [PubMed] [Google Scholar]
- Mansour A, Thompson RC, Akil H, Watson SJ. Delta opioid receptor mRNA distribution in the brain: comparison to delta receptor binding and proenkephalin mRNA. J Chem Neuroanat. 1993;6:351–362. doi: 10.1016/0891-0618(93)90010-2. [DOI] [PubMed] [Google Scholar]
- Mansour A, Fox CA, Thompson RC, Akil H, Watson SJ. mu-Opioid receptor mRNA expression in the rat CNS: comparison to mu-receptor binding. Brain Res. 1994a;643:245–265. doi: 10.1016/0006-8993(94)90031-0. [DOI] [PubMed] [Google Scholar]
- Mansour A, Fox CA, Burke S, Meng F, Thompson RC, Akil H, et al. Mu, delta, and kappa opioid receptor mRNA expression in the rat CNS: an in situ hybridization study. J Comp Neurol. 1994b;350:412–438. doi: 10.1002/cne.903500307. [DOI] [PubMed] [Google Scholar]
- Mansour A, Fox CA, Akil H, Watson SJ. Opioid-receptor mRNA expression in the rat CNS: anatomical and functional implications. Trends Neurosci. 1995;18:22–29. doi: 10.1016/0166-2236(95)93946-u. [DOI] [PubMed] [Google Scholar]
- Marinelli PW, Bai L, Quirion R, Gianoulakis C. A microdialysis profile of Met-enkephalin release in the rat nucleus accumbens following alcohol administration. Alcohol Clin Exp Res. 2005;29:1821–1828. doi: 10.1097/01.alc.0000183008.62955.2e. [DOI] [PubMed] [Google Scholar]
- Mason BJ, Salvato FR, Williams LD, Ritvo EC, Cutler RB. A double-blind, placebo-controlled study of oral nalmefene for alcohol dependence. Arch Gen Psychiatry. 1999;56:719–724. doi: 10.1001/archpsyc.56.8.719. [DOI] [PubMed] [Google Scholar]
- Mathieu AM, Caboche J, Besson MJ. Distribution of preproenkephalin, preprotachykinin A, and preprodynorphin mRNAs in the rat nucleus accumbens: effect of repeated administration of nicotine. Synapse. 1996;23:94–106. doi: 10.1002/(SICI)1098-2396(199606)23:2<94::AID-SYN5>3.0.CO;2-B. [DOI] [PubMed] [Google Scholar]
- Mathieu-Kia AM, Besson MJ. Repeated administration of cocaine, nicotine and ethanol: effects on preprodynorphin, preprotachykinin A and preproenkephalin mRNA expression in the dorsal and the ventral striatum of the rat. Brain Res Mol Brain Res. 1998;54:141–151. doi: 10.1016/s0169-328x(97)00338-0. [DOI] [PubMed] [Google Scholar]
- Mathon DS, Lesscher HM, Gerrits MA, Kamal A, Pintar JE, Schuller AG, et al. Increased gabaergic input to ventral tegmental area dopaminergic neurons associated with decreased cocaine reinforcement in mu-opioid receptor knockout mice. Neuroscience. 2005;130:359–367. doi: 10.1016/j.neuroscience.2004.10.002. [DOI] [PubMed] [Google Scholar]
- McCarthy MJ, Zhang H, Neff NH, Hadjiconstantinou M. Nicotine withdrawal and kappa-opioid receptors. Psychopharmacology (Berl) 2010;210:221–229. doi: 10.1007/s00213-009-1674-5. [DOI] [PubMed] [Google Scholar]
- McCarthy MJ, Zhang H, Neff NH, Hadjiconstantinou M. Desensitization of delta-opioid receptors in nucleus accumbens during nicotine withdrawal. Psychopharmacology (Berl) 2011;213:735–744. doi: 10.1007/s00213-010-2028-z. [DOI] [PubMed] [Google Scholar]
- Mello NK, Mendelson JH, Sellers ML, Kuehnle JC. Effects of heroin self-administration on cigarette smoking. Psychopharmacology (Berl) 1980;67:45–52. doi: 10.1007/BF00427594. [DOI] [PubMed] [Google Scholar]
- Mello NK, Lukas SE, Mendelson JH. Buprenorphine effects on cigarette smoking. Psychopharmacology (Berl) 1985;86:417–425. doi: 10.1007/BF00427902. [DOI] [PubMed] [Google Scholar]
- Mello NK, Mendelson JH, Bree MP, Lukas SE. Buprenorphine suppresses cocaine self-administration by rhesus monkeys. Science. 1989;245:859–862. doi: 10.1126/science.2772637. [DOI] [PubMed] [Google Scholar]
- Mendelson JH, Teoh SK, Mello NK, Ellingboe J. Buprenorphine attenuates the effects of cocaine on adrenocorticotropin (ACTH) secretion and mood states in man. Neuropsychopharmacology. 1992;7:157–162. [PubMed] [Google Scholar]
- Mendez M, Morales-Mulia M. Ethanol exposure differentially alters pro-enkephalin mRNA expression in regions of the mesocorticolimbic system. Psychopharmacology (Berl) 2006;189:117–124. doi: 10.1007/s00213-006-0503-3. [DOI] [PubMed] [Google Scholar]
- Mendez M, Leriche M, Carlos Calva J. Acute ethanol administration transiently decreases [3H]-DAMGO binding to mu opioid receptors in the rat substantia nigra pars reticulata but not in the caudate-putamen. Neurosci Res. 2003;47:153–160. doi: 10.1016/s0168-0102(03)00188-3. [DOI] [PubMed] [Google Scholar]
- Mendez M, Barbosa-Luna IG, Perez-Luna JM, Cupo A, Oikawa J. Effects of acute ethanol administration on methionine-enkephalin expression and release in regions of the rat brain. Neuropeptides. 2010;44:413–420. doi: 10.1016/j.npep.2010.05.001. [DOI] [PubMed] [Google Scholar]
- Meng F, Xie GX, Thompson RC, Mansour A, Goldstein A, Watson SJ, et al. Cloning and pharmacological characterization of a rat kappa opioid receptor. Proc Natl Acad Sci U S A. 1993;90:9954–9958. doi: 10.1073/pnas.90.21.9954. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Menkens K, Bilsky EJ, Wild KD, Portoghese PS, Reid LD, Porreca F. Cocaine place preference is blocked by the delta-opioid receptor antagonist, naltrindole. Eur J Pharmacol. 1992;219:345–346. doi: 10.1016/0014-2999(92)90319-y. [DOI] [PubMed] [Google Scholar]
- Montoya ID, Gorelick DA, Preston KL, Schroeder JR, Umbricht A, Cheskin LJ, et al. Randomized trial of buprenorphine for treatment of concurrent opiate and cocaine dependence. Clin Pharmacol Ther. 2004;75:34–48. doi: 10.1016/j.clpt.2003.09.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mooney LJ, Nielsen S, Saxon A, Hillhouse M, Thomas C, Hasson A, et al. Cocaine use reduction with buprenorphine (CURB): rationale, design, and methodology. Contemp Clin Trials. 2013;34:196–204. doi: 10.1016/j.cct.2012.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morani AS, Schenk S, Prisinzano TE, Kivell BM. A single injection of a novel kappa opioid receptor agonist salvinorin A attenuates the expression of cocaine-induced behavioral sensitization in rats. Behav Pharmacol. 2012;23:162–170. doi: 10.1097/FBP.0b013e3283512c1e. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moratalla R, Xu M, Tonegawa S, Graybiel AM. Cellular responses to psychomotor stimulant and neuroleptic drugs are abnormal in mice lacking the D1 dopamine receptor. Proc Natl Acad Sci U S A. 1996;93:14928–14933. doi: 10.1073/pnas.93.25.14928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morris PL, Hopwood M, Whelan G, Gardiner J, Drummond E. Naltrexone for alcohol dependence: a randomized controlled trial. Addiction. 2001;96:1565–1573. doi: 10.1046/j.1360-0443.2001.961115654.x. [DOI] [PubMed] [Google Scholar]
- Mutschler NH, Stephen BJ, Teoh SK, Mendelson JH, Mello NK. An inpatient study of the effects of buprenorphine on cigarette smoking in men concurrently dependent on cocaine and opioids. Nicotine Tob Res. 2002;4:223–228. doi: 10.1080/14622200210124012. [DOI] [PubMed] [Google Scholar]
- Nemeth-Coslett R, Griffiths RR. Naloxone does not affect cigarette smoking. Psychopharmacology (Berl) 1986;89:261–264. doi: 10.1007/BF00174355. [DOI] [PubMed] [Google Scholar]
- Nestby P, Schoffelmeer AN, Homberg JR, Wardeh G, De Vries TJ, Mulder AH, et al. Bremazocine reduces unrestricted free-choice ethanol self-administration in rats without affecting sucrose preference. Psychopharmacology (Berl) 1999;142:309–317. doi: 10.1007/s002130050894. [DOI] [PubMed] [Google Scholar]
- Neumeyer JL, Gu XH, van Vliet LA, DeNunzio NJ, Rusovici DE, Cohen DJ, et al. Mixed kappa agonists and mu agonists/antagonists as potential pharmacotherapeutics for cocaine abuse: synthesis and opioid receptor binding affinity of N-substituted derivatives of morphinan. Bioorg Med Chem Lett. 2001;11:2735–2740. doi: 10.1016/s0960-894x(01)00543-1. [DOI] [PubMed] [Google Scholar]
- Nguyen AT, Marquez P, Hamid A, Kieffer B, Friedman TC, Lutfy K. The rewarding action of acute cocaine is reduced in beta-endorphin deficient but not in mu opioid receptor knockout mice. Eur J Pharmacol. 2012;686:50–54. doi: 10.1016/j.ejphar.2012.04.040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Noble F, Roques BP. Inhibitors of Enkephalin Catabolism. Totowa, NJ: Humana Press; 2003. [Google Scholar]
- O'Malley SS, Cooney JL, Krishnan-Sarin S, Dubin JA, McKee SA, Cooney NL, et al. A controlled trial of naltrexone augmentation of nicotine replacement therapy for smoking cessation. Arch Intern Med. 2006;166:667–674. doi: 10.1001/archinte.166.6.667. [DOI] [PubMed] [Google Scholar]
- Oslin DW, Berrettini W, Kranzler HR, Pettinati H, Gelernter J, Volpicelli JR, et al. A functional polymorphism of the mu-opioid receptor gene is associated with naltrexone response in alcohol-dependent patients. Neuropsychopharmacology. 2003;28:1546–1552. doi: 10.1038/sj.npp.1300219. [DOI] [PubMed] [Google Scholar]
- Oslin DW, Berrettini WH, O'Brien CP. Targeting treatments for alcohol dependence: the pharmacogenetics of naltrexone. Addict Biol. 2006;11:397–403. doi: 10.1111/j.1369-1600.2006.00036.x. [DOI] [PubMed] [Google Scholar]
- Parkes H, Sinclair JD. Reduction of alcohol drinking and upregulation of opioid receptors by oral naltrexone in AA rats. Alcohol. 2000;21:215–221. doi: 10.1016/s0741-8329(00)00091-4. [DOI] [PubMed] [Google Scholar]
- Pawson AJ, Sharman JL, Benson HE, Faccenda E, Alexander SP, Buneman OP, et al. NC-IUPHAR. The IUPHAR/BPS Guide to PHARMACOLOGY: an expert-driven knowledgebase of drug targets and their ligands. Nucl Acids Res. 2014;42(Database Issue):D1098–D1106. doi: 10.1093/nar/gkt1143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Perry CJ, McNally GP. A role for the ventral pallidum in context-induced and primed reinstatement of alcohol seeking. Eur J Neurosci. 2013a;38:2762–2773. doi: 10.1111/ejn.12283. [DOI] [PubMed] [Google Scholar]
- Perry CJ, McNally GP. mu-Opioid receptors in the nucleus accumbens shell mediate context-induced reinstatement (renewal) but not primed reinstatement of extinguished alcohol seeking. Behav Neurosci. 2013b;127:535–543. doi: 10.1037/a0032981. [DOI] [PubMed] [Google Scholar]
- Pert CB, Snyder SH. Opiate receptor: demonstration in nervous tissue. Science. 1973;179:1011–1014. doi: 10.1126/science.179.4077.1011. [DOI] [PubMed] [Google Scholar]
- Pettinati HM, Gastfriend DR, Dong Q, Kranzler HR, O'Malley SS. Effect of extended-release naltrexone (XR-NTX) on quality of life in alcohol-dependent patients. Alcohol Clin Exp Res. 2009;33:350–356. doi: 10.1111/j.1530-0277.2008.00843.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Placenza FM, Rajabi H, Stewart J. Effects of chronic buprenorphine treatment on levels of nucleus accumbens glutamate and on the expression of cocaine-induced behavioral sensitization in rats. Psychopharmacology (Berl) 2008;200:347–355. doi: 10.1007/s00213-008-1210-z. [DOI] [PubMed] [Google Scholar]
- Pomerleau OF, Fertig JB, Seyler LE, Jaffe J. Neuroendocrine reactivity to nicotine in smokers. Psychopharmacology (Berl) 1983;81:61–67. doi: 10.1007/BF00439275. [DOI] [PubMed] [Google Scholar]
- Potenza MN, Sofuoglu M, Carroll KM, Rounsaville BJ. Neuroscience of behavioral and pharmacological treatments for addictions. Neuron. 2011;69:695–712. doi: 10.1016/j.neuron.2011.02.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Przewlocka B, Lason W. Adaptive changes in the proenkephalin and D2 dopamine receptor mRNA expression after chronic cocaine in the nucleus accumbens and striatum of the rat. Eur Neuropsychopharmacol. 1995;5:465–469. doi: 10.1016/0924-977x(95)80005-m. [DOI] [PubMed] [Google Scholar]
- Przewlocka B, Lason W, Przewlocki R. Repeated ethanol differently affects opioid peptide biosynthesis in the rat pituitary. Neuroendocrinology. 1994;60:331–336. doi: 10.1159/000126766. [DOI] [PubMed] [Google Scholar]
- Przewlocka B, Turchan J, Lason W, Przewlocki R. Ethanol withdrawal enhances the prodynorphin system activity in the rat nucleus accumbens. Neurosci Lett. 1997;238:13–16. doi: 10.1016/s0304-3940(97)00829-x. [DOI] [PubMed] [Google Scholar]
- Rademacher DJ, Steinpreis RE. Effects of the selective mu(1)-opioid receptor antagonist, naloxonazine, on cocaine-induced conditioned place preference and locomotor behavior in rats. Neurosci Lett. 2002;332:159–162. doi: 10.1016/s0304-3940(02)00950-3. [DOI] [PubMed] [Google Scholar]
- Ramsey NF, Gerrits MA, Van Ree JM. Naltrexone affects cocaine self-administration in naive rats through the ventral tegmental area rather than dopaminergic target regions. Eur Neuropsychopharmacol. 1999;9:93–99. doi: 10.1016/s0924-977x(98)00009-1. [DOI] [PubMed] [Google Scholar]
- Rasmussen DD. Effects of chronic nicotine treatment and withdrawal on hypothalamic proopiomelanocortin gene expression and neuroendocrine regulation. Psychoneuroendocrinology. 1998;23:245–259. doi: 10.1016/s0306-4530(98)00003-1. [DOI] [PubMed] [Google Scholar]
- Raynor K, Kong H, Chen Y, Yasuda K, Yu L, Bell GI, et al. Pharmacological characterization of the cloned kappa-, delta-, and mu-opioid receptors. Mol Pharmacol. 1994;45:330–334. [PubMed] [Google Scholar]
- Reid LD, Hunter GA. Morphine and naloxone modulate intake of ethanol. Alcohol. 1984;1:33–37. doi: 10.1016/0741-8329(84)90033-8. [DOI] [PubMed] [Google Scholar]
- Reid LD, Glick SD, Menkens KA, French ED, Bilsky EJ, Porreca F. Cocaine self-administration and naltrindole, a delta-selective opioid antagonist. Neuroreport. 1995;6:1409–1412. doi: 10.1097/00001756-199507100-00012. [DOI] [PubMed] [Google Scholar]
- Roberts AJ, McDonald JS, Heyser CJ, Kieffer BL, Matthes HW, Koob GF, et al. mu-Opioid receptor knockout mice do not self-administer alcohol. J Pharmacol Exp Ther. 2000;293:1002–1008. [PubMed] [Google Scholar]
- Roberts AJ, Gold LH, Polis I, McDonald JS, Filliol D, Kieffer BL, et al. Increased ethanol self-administration in delta-opioid receptor knockout mice. Alcohol Clin Exp Res. 2001;25:1249–1256. [PubMed] [Google Scholar]
- Rohsenow DJ, Monti PM, Hutchison KE, Swift RM, MacKinnon SV, Sirota AD, et al. High-dose transdermal nicotine and naltrexone: effects on nicotine withdrawal, urges, smoking, and effects of smoking. Exp Clin Psychopharmacol. 2007;15:81–92. doi: 10.1037/1064-1297.15.1.81. [DOI] [PubMed] [Google Scholar]
- Romualdi P, D'Addario C, Ferri S, Cox BM, Izenwasser S. Chronic GBR 12909 administration differentially alters prodynorphin gene expression compared to cocaine. Eur J Pharmacol. 2001;413:207–212. doi: 10.1016/s0014-2999(01)00776-2. [DOI] [PubMed] [Google Scholar]
- Roques BP, Noble F. Association of enkephalin catabolism inhibitors and CCK-B antagonists: a potential use in the management of pain and opioid addiction. Neurochem Res. 1996;21:1397–1410. doi: 10.1007/BF02532381. [DOI] [PubMed] [Google Scholar]
- Rorick-Kehn LM, Witkin JM, Statnick MA, Eberle EL, McKinzie JH, Kahl SD, et al. LY2456302 is a novel, potent, orally-bioavailable small molecule kappa-selective antagonist with activity in animal models predictive of efficacy in mood and addictive disorders. Neuropharmacology. 2014;77:131–144. doi: 10.1016/j.neuropharm.2013.09.021. [DOI] [PubMed] [Google Scholar]
- Rosecrans JA, Hendry JS, Hong JS. Biphasic effects of chronic nicotine treatment on hypothalamic immunoreactive beta-endorphin in the mouse. Pharmacol Biochem Behav. 1985;23:141–143. doi: 10.1016/0091-3057(85)90141-8. [DOI] [PubMed] [Google Scholar]
- Roth-Deri I, Zangen A, Aleli M, Goelman RG, Pelled G, Nakash R, et al. Effect of experimenter-delivered and self-administered cocaine on extracellular beta-endorphin levels in the nucleus accumbens. J Neurochem. 2003;84:930–938. doi: 10.1046/j.1471-4159.2003.01584.x. [DOI] [PubMed] [Google Scholar]
- Roth-Deri I, Green-Sadan T, Yadid G. Beta-endorphin and drug-induced reward and reinforcement. Prog Neurobiol. 2008;86:1–21. doi: 10.1016/j.pneurobio.2008.06.003. [DOI] [PubMed] [Google Scholar]
- Saland LC, Hastings CM, Abeyta A, Chavez JB. Chronic ethanol modulates delta and mu-opioid receptor expression in rat CNS: immunohistochemical analysis with quantitative confocal microscopy. Neurosci Lett. 2005;381:163–168. doi: 10.1016/j.neulet.2005.02.016. [DOI] [PubMed] [Google Scholar]
- Schroeder JA, Hummel M, Simpson AD, Sheikh R, Soderman AR, Unterwald EM. A role for mu opioid receptors in cocaine-induced activity, sensitization, and reward in the rat. Psychopharmacology (Berl) 2007;195:265–272. doi: 10.1007/s00213-007-0883-z. [DOI] [PubMed] [Google Scholar]
- Schulz R, Wuster M, Duka T, Herz A. Acute and chronic ethanol treatment changes endorphin levels in brain and pituitary. Psychopharmacology (Berl) 1980;68:221–227. doi: 10.1007/BF00428107. [DOI] [PubMed] [Google Scholar]
- Scott DJ, Domino EF, Heitzeg MM, Koeppe RA, Ni L, Guthrie S, et al. Smoking modulation of mu-opioid and dopamine D2 receptor-mediated neurotransmission in humans. Neuropsychopharmacology. 2007;32:450–457. doi: 10.1038/sj.npp.1301238. [DOI] [PubMed] [Google Scholar]
- Seizinger BR, Bovermann K, Maysinger D, Hollt V, Herz A. Differential effects of acute and chronic ethanol treatment on particular opioid peptide systems in discrete regions of rat brain and pituitary. Pharmacol Biochem Behav. 1983;18(Suppl. 1):361–369. doi: 10.1016/0091-3057(83)90200-9. [DOI] [PubMed] [Google Scholar]
- Seyler LE, Jr, Pomerleau OF, Fertig JB, Hunt D, Parker K. Pituitary hormone response to cigarette smoking. Pharmacol Biochem Behav. 1986;24:159–162. doi: 10.1016/0091-3057(86)90062-6. [DOI] [PubMed] [Google Scholar]
- Simon EJ, Hiller JM, Edelman I. Stereospecific binding of the potent narcotic analgesic (3H) Etorphine to rat-brain homogenate. Proc Natl Acad Sci U S A. 1973;70:1947–1949. doi: 10.1073/pnas.70.7.1947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sivam SP. Cocaine selectively increases striatonigral dynorphin levels by a dopaminergic mechanism. J Pharmacol Exp Ther. 1989;250:818–824. [PubMed] [Google Scholar]
- Smiley PL, Johnson M, Bush L, Gibb JW, Hanson GR. Effects of cocaine on extrapyramidal and limbic dynorphin systems. J Pharmacol Exp Ther. 1990;253:938–943. [PubMed] [Google Scholar]
- Sorge RE, Stewart J. The effects of chronic buprenorphine on intake of heroin and cocaine in rats and its effects on nucleus accumbens dopamine levels during self-administration. Psychopharmacology (Berl) 2006;188:28–41. doi: 10.1007/s00213-006-0485-1. [DOI] [PubMed] [Google Scholar]
- Sorge RE, Rajabi H, Stewart J. Rats maintained chronically on buprenorphine show reduced heroin and cocaine seeking in tests of extinction and drug-induced reinstatement. Neuropsychopharmacology. 2005;30:1681–1692. doi: 10.1038/sj.npp.1300712. [DOI] [PubMed] [Google Scholar]
- Spangler R, Ho A, Zhou Y, Maggos CE, Yuferov V, Kreek MJ. Regulation of kappa opioid receptor mRNA in the rat brain by ‘binge’ pattern cocaine administration and correlation with preprodynorphin mRNA. Brain Res Mol Brain Res. 1996a;38:71–76. doi: 10.1016/0169-328x(95)00319-n. [DOI] [PubMed] [Google Scholar]
- Spangler R, Zhou Y, Maggos CE, Zlobin A, Ho A, Kreek MJ. Dopamine antagonist and ‘binge’ cocaine effects on rat opioid and dopamine transporter mRNAs. Neuroreport. 1996b;7:2196–2200. doi: 10.1097/00001756-199609020-00028. [DOI] [PubMed] [Google Scholar]
- Staley JK, Rothman RB, Rice KC, Partilla J, Mash DC. Kappa2 opioid receptors in limbic areas of the human brain are upregulated by cocaine in fatal overdose victims. J Neurosci. 1997;17:8225–8233. doi: 10.1523/JNEUROSCI.17-21-08225.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Steiner H, Gerfen CR. Cocaine-induced c-fos messenger RNA is inversely related to dynorphin expression in striatum. J Neurosci. 1993;13:5066–5081. doi: 10.1523/JNEUROSCI.13-12-05066.1993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Strain EC, Stitzer ML, Liebson IA, Bigelow GE. Buprenorphine versus methadone in the treatment of opioid-dependent cocaine users. Psychopharmacology (Berl) 1994;116:401–406. doi: 10.1007/BF02247469. [DOI] [PubMed] [Google Scholar]
- Stromberg MF, Volpicelli JR, O'Brien CP. Effects of naltrexone administered repeatedly across 30 or 60 days on ethanol consumption using a limited access procedure in the rat. Alcohol Clin Exp Res. 1998;22:2186–2191. [PubMed] [Google Scholar]
- Stromberg MF, Rukstalis MR, Mackler SA, Volpicelli JR, O'Brien CP. A comparison of the effects of 6-beta naltrexol and naltrexone on the consumption of ethanol or sucrose using a limited-access procedure in rats. Pharmacol Biochem Behav. 2002;72:483–490. doi: 10.1016/s0091-3057(02)00721-9. [DOI] [PubMed] [Google Scholar]
- Sturgess JE, George TP, Kennedy JL, Heinz A, Muller DJ. Pharmacogenetics of alcohol, nicotine and drug addiction treatments. Addict Biol. 2011;16:357–376. doi: 10.1111/j.1369-1600.2010.00287.x. [DOI] [PubMed] [Google Scholar]
- Suzuki T, Shiozaki Y, Masukawa Y, Misawa M, Nagase H. The role of mu- and kappa-opioid receptors in cocaine-induced conditioned place preference. Jpn J Pharmacol. 1992;58:435–442. doi: 10.1254/jjp.58.435. [DOI] [PubMed] [Google Scholar]
- Suzuki T, Mori T, Tsuji M, Misawa M, Nagase H. The role of delta-opioid receptor subtypes in cocaine- and methamphetamine-induced place preferences. Life Sci. 1994;55:PL339–PL344. doi: 10.1016/0024-3205(94)00774-8. [DOI] [PubMed] [Google Scholar]
- Svensson P, Hurd YL. Specific reductions of striatal prodynorphin and D1 dopamine receptor messenger RNAs during cocaine abstinence. Brain Res Mol Brain Res. 1998;56:162–168. doi: 10.1016/s0169-328x(98)00041-2. [DOI] [PubMed] [Google Scholar]
- Terenius L. Stereospecific interaction between narcotic analgesics and a synaptic plasm a membrane fraction of rat cerebral cortex. Acta Pharmacol Toxicol (Copenh) 1973;32:317–320. doi: 10.1111/j.1600-0773.1973.tb01477.x. [DOI] [PubMed] [Google Scholar]
- Thompson RC, Mansour A, Akil H, Watson SJ. Cloning and pharmacological characterization of a rat mu opioid receptor. Neuron. 1993;11:903–913. doi: 10.1016/0896-6273(93)90120-g. [DOI] [PubMed] [Google Scholar]
- Thorsell A. The mu-opioid receptor and treatment response to naltrexone. Alcohol Alcohol. 2013;48:402–408. doi: 10.1093/alcalc/agt030. [DOI] [PubMed] [Google Scholar]
- Toll BA, White M, Wu R, Meandzija B, Jatlow P, Makuch R, et al. Low-dose naltrexone augmentation of nicotine replacement for smoking cessation with reduced weight gain: a randomized trial. Drug Alcohol Depend. 2010;111:200–206. doi: 10.1016/j.drugalcdep.2010.04.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tomasiewicz HC, Todtenkopf MS, Chartoff EH, Cohen BM, Carlezon WA., Jr The kappa-opioid agonist U69,593 blocks cocaine-induced enhancement of brain stimulation reward. Biol Psychiatry. 2008;64:982–988. doi: 10.1016/j.biopsych.2008.05.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Torres G, Horowitz JM. Drugs of abuse and brain gene expression. Psychosom Med. 1999;61:630–650. doi: 10.1097/00006842-199909000-00007. [DOI] [PubMed] [Google Scholar]
- Trigo JM, Zimmer A, Maldonado R. Nicotine anxiogenic and rewarding effects are decreased in mice lacking beta-endorphin. Neuropharmacology. 2009;56:1147–1153. doi: 10.1016/j.neuropharm.2009.03.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turchan J, Przewlocka B, Lason W, Przewlocki R. Effects of repeated psychostimulant administration on the prodynorphin system activity and kappa opioid receptor density in the rat brain. Neuroscience. 1998;85:1051–1059. doi: 10.1016/s0306-4522(97)00639-8. [DOI] [PubMed] [Google Scholar]
- Tzschentke TM. Measuring reward with the conditioned place preference (CPP) paradigm: update of the last decade. Addict Biol. 2007;12:227–462. doi: 10.1111/j.1369-1600.2007.00070.x. [DOI] [PubMed] [Google Scholar]
- Vanderschuren LJ, Kalivas PW. Alterations in dopaminergic and glutamatergic transmission in the induction and expression of behavioral sensitization: a critical review of preclinical studies. Psychopharmacology (Berl) 2000;151:99–120. doi: 10.1007/s002130000493. [DOI] [PubMed] [Google Scholar]
- Vihavainen T, Piltonen M, Tuominen RK, Korpi ER, Ahtee L. Morphine-nicotine interaction in conditioned place preference in mice after chronic nicotine exposure. Eur J Pharmacol. 2008;587:169–174. doi: 10.1016/j.ejphar.2008.03.028. [DOI] [PubMed] [Google Scholar]
- Volkow ND, Skolnick P. New medications for substance use disorders: challenges and opportunities. Neuropsychopharmacology. 2012;37:290–292. doi: 10.1038/npp.2011.84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Vries TJ, Babovic-Vuksanovic D, Elmer G, Shippenberg TS. Lack of involvement of delta-opioid receptors in mediating the rewarding effects of cocaine. Psychopharmacology (Berl) 1995;120:442–448. doi: 10.1007/BF02245816. [DOI] [PubMed] [Google Scholar]
- Walker BM, Koob GF. Pharmacological evidence for a motivational role of kappa-opioid systems in ethanol dependence. Neuropsychopharmacology. 2008;33:643–652. doi: 10.1038/sj.npp.1301438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Walters CL, Cleck JN, Kuo YC, Blendy JA. Mu-opioid receptor and CREB activation are required for nicotine reward. Neuron. 2005;46:933–943. doi: 10.1016/j.neuron.2005.05.005. [DOI] [PubMed] [Google Scholar]
- Wang D, Sun X, Sadee W. Different effects of opioid antagonists on mu-, delta-, and kappa-opioid receptors with and without agonist pretreatment. J Pharmacol Exp Ther. 2007;321:544–552. doi: 10.1124/jpet.106.118810. [DOI] [PubMed] [Google Scholar]
- Ward SJ, Roberts DC. Microinjection of the delta-opioid receptor selective antagonist naltrindole 5′-isothiocyanate site specifically affects cocaine self-administration in rats responding under a progressive ratio schedule of reinforcement. Behav Brain Res. 2007;182:140–144. doi: 10.1016/j.bbr.2007.05.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wee S, Koob GF. The role of the dynorphin-kappa opioid system in the reinforcing effects of drugs of abuse. Psychopharmacology (Berl) 2010;210:121–135. doi: 10.1007/s00213-010-1825-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wee S, Vendruscolo LF, Misra KK, Schlosburg JE, Koob GF. A combination of buprenorphine and naltrexone blocks compulsive cocaine intake in rodents without producing dependence. Sci Transl Med. 2012;4:146ra110. doi: 10.1126/scitranslmed.3003948. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Weerts EM, Wand GS, Kuwabara H, Xu X, Frost JJ, Wong DF, et al. Association of smoking with mu-opioid receptor availability before and during naltrexone blockade in alcohol-dependent subjects. Addict Biol. 2014;19:733–742. doi: 10.1111/adb.12022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wewers ME, Dhatt R, Tejwani GA. Naltrexone administration affects ad libitum smoking behavior. Psychopharmacology (Berl) 1998;140:185–190. doi: 10.1007/s002130050756. [DOI] [PubMed] [Google Scholar]
- Wewers ME, Dhatt RK, Snively TA, Tejwani GA. The effect of chronic administration of nicotine on antinociception, opioid receptor binding and met-enkephalin levels in rats. Brain Res. 1999;822:107–113. doi: 10.1016/s0006-8993(99)01095-1. [DOI] [PubMed] [Google Scholar]
- Williams KL, Woods JH. Oral ethanol-reinforced responding in rhesus monkeys: effects of opioid antagonists selective for the mu-, kappa-, or delta-receptor. Alcohol Clin Exp Res. 1998;22:1634–1639. doi: 10.1111/j.1530-0277.1998.tb03960.x. [DOI] [PubMed] [Google Scholar]
- Williams TM, Davies SJ, Taylor LG, Daglish MR, Hammers A, Brooks DJ, et al. Brain opioid receptor binding in early abstinence from alcohol dependence and relationship to craving: an [11C]diprenorphine PET study. Eur Neuropsychopharmacol. 2009;19:740–748. doi: 10.1016/j.euroneuro.2009.06.007. [DOI] [PubMed] [Google Scholar]
- Wong CJ, Witcher J, Mallinckrodt C, Dean RA, Anton RF, Chen Y, et al. A phase 2, placebo-controlled study of the opioid receptor antagonist LY2196044 for the treatment of alcohol dependence. Alcohol Clin Exp Res. 2014;38:511–520. doi: 10.1111/acer.12257. [DOI] [PubMed] [Google Scholar]
- Wong GY, Wolter TD, Croghan GA, Croghan IT, Offord KP, Hurt RD. A randomized trial of naltrexone for smoking cessation. Addiction. 1999;94:1227–1237. doi: 10.1046/j.1360-0443.1999.948122713.x. [DOI] [PubMed] [Google Scholar]
- Yasuda K, Raynor K, Kong H, Breder CD, Takeda J, Reisine T, et al. Cloning and functional comparison of kappa and delta opioid receptors from mouse brain. Proc Natl Acad Sci U S A. 1993;90:6736–6740. doi: 10.1073/pnas.90.14.6736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yoo JH, Kitchen I, Bailey A. The endogenous opioid system in cocaine addiction: what lessons have opioid peptide and receptor knockout mice taught us? Br J Pharmacol. 2012;166:1993–2014. doi: 10.1111/j.1476-5381.2012.01952.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yuferov V, Zhou Y, Spangler R, Maggos CE, Ho A, Kreek MJ. Acute ‘binge’ cocaine increases mu-opioid receptor mRNA levels in areas of the rat mesolimbic mesocortical dopamine system. Brain Res Bull. 1999;48:109–112. doi: 10.1016/s0361-9230(98)00155-5. [DOI] [PubMed] [Google Scholar]
- Zarrindast MR, Faraji N, Rostami P, Sahraei H, Ghoshouni H. Cross-tolerance between morphine- and nicotine-induced conditioned place preference in mice. Pharmacol Biochem Behav. 2003;74:363–369. doi: 10.1016/s0091-3057(02)01002-x. [DOI] [PubMed] [Google Scholar]
- Zhang Y, Schlussman SD, Butelman ER, Ho A, Kreek MJ. Effects of withdrawal from chronic escalating-dose binge cocaine on conditioned place preference to cocaine and striatal preproenkephalin mRNA in C57BL/6J mice. Neuropharmacology. 2012;63:322–329. doi: 10.1016/j.neuropharm.2012.03.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhou Y, Colombo G, Niikura K, Carai MA, Femenia T, Garcia-Gutierrez MS, et al. Voluntary alcohol drinking enhances proopiomelanocortin gene expression in nucleus accumbens shell and hypothalamus of Sardinian alcohol-preferring rats. Alcohol Clin Exp Res. 2013;37(Suppl. 1):E131–E140. doi: 10.1111/j.1530-0277.2012.01867.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ziolkowska B, Stefanski R, Mierzejewski P, Zapart G, Kostowski W, Przewlocki R. Contingency does not contribute to the effects of cocaine self-administration on prodynorphin and proenkephalin gene expression in the rat forebrain. Brain Res. 2006;1069:1–9. doi: 10.1016/j.brainres.2005.11.042. [DOI] [PubMed] [Google Scholar]
- Zirakzadeh A, Shuman C, Stauter E, Hays JT, Ebbert JO. Cigarette smoking in methadone maintained patients: an up-to-date review. Curr Drug Abuse Rev. 2013;6:77–84. doi: 10.2174/1874473711306010009. [DOI] [PubMed] [Google Scholar]
- Zubieta JK, Gorelick DA, Stauffer R, Ravert HT, Dannals RF, Frost JJ. Increased mu opioid receptor binding detected by PET in cocaine-dependent men is associated with cocaine craving. Nat Med. 1996;2:1225–1229. doi: 10.1038/nm1196-1225. [DOI] [PubMed] [Google Scholar]